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.
Keywords: Myeloproliferative neoplasms, myelofibrosis, therapeutic approaches
Introduction
The classical BCR-ABL1-negative myeloproliferative neoplasms (MPNs) (denoted here thereafter MPNs) include three different disorders, essential thrombocythemia (ET), polycythemia vera (PV) and primary myelofibrosis (PMF). They may be seen as 3 stages of the same disorder as they are all driven by constitutive activation of JAK2 by mutations in 3 genes1,2. Disease phenotype and prognosis are related to the precise disease driver mutation along with its genetic status (heterozygous or homozygous), but also to the presence of other acquired “clonal” driver mutations. The latter are also associated with other myeloid malignancies and clonal hematopoiesis, but they do not per se induce MPN. The combination of acquired mutations, their number and order of acquisition play a central role in the phenotype and prognosis1–3. In addition, inherited genetic factors and environmental cues such as inflammation and iron metabolism dysregulation may also impact phenotype and prognosis1.
PMF is the most severe MPN. Diagnosis is based on bone marrow histology with the presence of megakaryocyte (MK) proliferation and marrow fibrosis (grade 2–3) associated with osteosclerosis in advanced cases4,5. Secondary MF is preceded by an ET or a PV. An overt PMF can be preceded by an early-PMF (pre-fibrosis) that shares many features with ET, but differs by the presence of a splenomegaly, increased bone marrow cellularity with dysplastic MK proliferation and eventually low-grade marrow fibrosis (grade<2). Most PMFs have disease driver mutations, except a subgroup denoted as triple negative PMF5,6.
The severity of the disease is related to the risk of leukemic transformation (14–25%), severe cytopenia, thromboembolic, hemorrhagic and infectious complications, cardiovascular disorders and cachexia.
There is no curative treatment of MF except allogeneic hematopoietic stem cell (HSC) transplantation, which remains risky, despite major progress in its management. Thus, to progress in the therapy of PMF, it is crucial to better understand the precise molecular bases of MPN and MF development.
MF and mutations
Disease driver mutations
MPN MF can be induced by 3 disease driver mutations. JAK2V617F is the predominant mutation present in around 55%–60% PMF1,2. JAK2V617F is a gain of function (GOF) mutation not located in the kinase domain, but in the pseudo-kinase (PK) domain. The V617F mutation activates the kinase domain by dimerization of the mutated PK domains, stabilizing a dimer state and removing negative regulation on the kinase domain7. The mutation can be heterozygous, or homozygous. JAK2V617F can induce constitutive signaling downstream of the three main “myeloid” homodimeric receptors (EPOR, G-CSFR, MPL), explaining that it may induce the three diseases. The 3 main signaling pathways include the STAT activation (STAT1, STAT3 and STAT5, according to the receptors), the PI3K/AKT/mTOR and the RAS/MAPK pathways. JAK2V617F not only induces proliferation of hematopoietic stem cell progenitor (HSCP) cells but gives a strong proliferative advantage to maturing precursors. It also activates or primes mature cells, inducing an inflammatory response, mainly through STAT3 activation and favoring thrombosis. Phylogenic reconstitution of the clone history in JAK2V617F MPN has shown that the mutation arises decades before disease development or in utero, giving the possibility of an early therapeutic approach8.
CALR mutations (mut) are the predominant disease driver mutation in JAK2 and MPL wild type (WT) PMF1,2. All described mutations are deletions/insertions in the exon 9, inducing a frameshift (-1/+2), creating a new C-terminus with positively charged and hydrophobic residues and lacking the KDEL endoplasmic reticulum (ER) retention signal. The two most frequent mutations are del52 (type-1) and ins5 (type-2), del52 mutations being enriched in PMF in comparison to ET9. CALR is a chaperone of the ER that plays a major role in the quality control of secreted glycoproteins and in calcium metabolism. The CALRmut, thanks to their C-terminus, specifically bind to N-glycosylated MPL via the lectin domain10–12. The CALRmut/MPL complex traffics to the cell surface; CALRmut are oligomers that dimerize MPL leading to persistent JAK2 activation13,14. CALRmut induce cell signaling at the cell surface via MPL or by being secreted and behaving as rogue cytokines that secondary bind to the immature sugars of MPL only present in the clone and by oligomerizing with endogenous CALRmut15. Thus, CALRmut can be directly targeted by different approaches. CALRmut may also exert effects via impaired calcium ER retention and subsequent activation of an ER stress response16,17. While CALRmut MPN develop on average nearly a decade before JAK2V617F MPN, CALRmut are acquired later in adult life, as inferred by mathematical modeling18. However, in a case of monozygotic twins it was shown that the mutation was acquired during fetal life19.
GOF MPL mutations are much rarer (around 5%). Mutations occur either in the cytosolic juxta-membrane domain at W515, usually W515L/K, or in the transmembrane domain, MPLS505N, both types leading to MPL dimerization and TPO independent megakaryopoiesis1,2,20. Other mutations can be found all along the MPL sequence. They are much rarer and exert weak GOF20.
“Clonal” driver mutations
In 80% of PMF, other “clonal” driver mutations are found that impact disease and prognosis. ASXL1, EZH2, SRSF2 and IDH1/IDH2 mutations are associated with an adverse prognosis5. Other mutations such as in N-RAS, K-RAS, CBL and other splicing genes induce also a worsened prognosis5. The role of these mutations in prognosis may also depend on the disease driver mutations. For example, the prognosis of CALR type-1 mutations is not altered by an ASXL1 mutation.
The role of TP53 mutations in prognosis is controversial but may be important in the choice of therapy21.
PMF is a heterogeneous disease concerning its prognosis due to the complex molecular alterations. Scores have been developed to stratify the prognosis and improve the choice of therapy based on clinical data, very high risk cytogenetic abnormalities (-7, i(17q), inv(3)/3q21, 12p-/12p11.2, 11q-/11q23, or other autosomal trisomies not including +8/+9) and high-molecular-risk mutations (HMR) (ASXL1, SRSF2, EZH2, IDH1 and U2AF1Q157)4,5.
Physiopathology of the marrow fibrosis
Marrow fibrosis (myelofibrosis) per se is characterized by the accumulation of extracellular matrix (ECM) fibers, namely collagen, that disorganizes the bone marrow environment and impairs hematopoiesis. It starts with the formation of a loose network of reticular fibers made of collagen III (grade-1) and then progresses with the accumulation of fibers composed of collagen I (grade-2) that will further accumulate with the appearance of an osteosclerosis (grade-3)6.
Marrow fibrosis can be associated with very different pathologies, such as auto-immune disorders, cancer metastases, lymphoma and myeloid malignancies. PMF may progress in two stages: one with a major myeloproliferation and the second called “myelodepletive” characterized by cytopenia22. PMF is associated with important extra-medullary hematopoiesis (EMH), more particularly in the spleen. The marrow fibrosis is usually considered to be reactive, as consequence of a cross-talk between the clonal disorder and stromal cells, which secrete ECM. ECM is also regulated by MKs that synthesize different types of collagen and extra-domain-A fibronectin and lysyl-oxidase, an enzyme playing an important role in ECM organization23,24.
TGF-β1 plays a central role in the development of all types of fibrosis and appears indispensable for marrow fibrosis development in mouse models25,26. It acts on mesenchymal stem cells (MSC) by inducing their proliferation and programming them to myofibroblasts and osteoblasts. TGF-β1 is secreted as an inactive latent form in a complex with TGF-β propeptide also called latency-associated protein (LAP), and latent TGF-β binding protein (LTBP). TGF-β1 is activated by different mechanisms including the interaction with αv integrins or the ECM such as thrombospondin 1 or the production of reactive oxygen species (ROS)25. In MF patients, the latent and active TGF-β1 levels are increased in bone marrow and plasma. The mechanism of TGF-β1 activation in MF is not clearly established and could be either related to the ROS level or the presentation of latent TGF-β1 on the cell surface by GARP through LAP binding to be subsequently activated by integrins. Initially it was found that GARP is only expressed on immune cells, but it is also present on the membrane of MKs/platelets as well as of some stromal cells27,28.
The other important determinant is the role of inflammation, more particularly of pro-fibrotic cytokines. The development of inflammation is an early event in the development of MPN. It has been shown that numerous plasma pro-inflammatory cytokines, such as IL8, IL12 and IL15 are predictive of the PMF prognosis5. The hematopoietic cells of the MPN as well as hematopoietic cells not belonging to the clone and non-hematopoietic cells are involved in inflammation29. It has been underscored that stromal cell inflammation may precede the development of the disease30. Pro-inflammatory cytokines may act by: (a) promoting a clonal advantage for cytokines (IL1β, IL8, TNFα and IL6)31; (b) inducing ROS release that may activate the latent TGF-β1; (c) reprogramming the Gli+ Lep+ MSC to myofibroblasts (IL1β and IL6 and alarmins S1008A/S1009A);32, and (d) acting on TGF-β1 synthesis and activation (IL13)28. Overall, the JAK/STAT3 and the NFκB pathways are important in the synthesis of pro-inflammatory cytokines and their effects29,33.
MKs appear to be a key cell determinant in marrow fibrosis development, as suggested by their close localization with collagen fibers34. They are the most important source of TGF-β1 in the bone marrow with a latent form that can be presented at the cell surface. In addition, MKs synthesize pro-inflammatory cytokines, chemokines and pro-angiogenic factors as well as ECM. The secretion of TGF-β1 and other pro-inflammatory cytokines, which are packaged in α-granules, is probably favored by the dysmegakaryopoiesis and the activation of MKs, as shown by the cell surface expression of P-selectin associated with an increased emperipolesis35.
These last years, the role of monocytes in marrow fibrosis development has been highlighted. Indeed, monocytes can release a lot of pro-inflammatory cytokines. In addition, SLAMF7 monocytes can differentiate to fibrocytes that are also considered as important mediators of marrow fibrosis in the JAK2V617F mouse model36,37. In this case, the marrow fibrosis will not only be a reactive process, but may directly derive from the clone. Recently, it has been suggested that Tregs play a central role in TGF-β1 production and activation and may also limit the CD8 T cell immune reaction against the clone27. Mast cells may also be involved in TGF-β1 activation by IL13 secretion28.
It must be underscored that an increased level of TPO in the mouse model called TPOhigh or the administration of MPL agonist, such as romiplostim, in mouse and in non-human primates leads to the development of a severe marrow fibrosis38. Similarly, in humans, administration of romiplostim may induce a moderate marrow fibrosis, which regresses at the arrest of the treatment39. In addition, the MPLW515L retroviral mouse model and MPLS504N knock in (KI) mice (human MPLS505N) develop a rapid marrow fibrosis40,41. Moreover, the genetic reversion of Jak2V617F to Jak2 WT in Jak2V617F KI mice induces a marrow fibrosis regression42. All these results demonstrate that a very strong activation of the TPO/MPL/JAK2 pathway is sufficient to induce the marrow fibrosis development and a pro-inflammatory state.
Presently, none of the pharmaceutical approaches have really improved the prognosis of high-risk MF4,43–45. Thus, besides JAK2 inhibitors and IFNα, many new therapies are in development, targeting different cellular processes, such as apoptosis, cell cycle, epigenetic regulators, signaling pathways, telomerase, or directly the leukemic stem cell (LSC). Many of these therapies are developed in association with JAK2 inhibitors with the risk of increasing hematological toxicity. The two principal goals of these new therapies are to: (a) significantly impact the clonal disorder and thus to revert the MF and to increase patient survival, and (b) provide alternative therapies in case of JAK2 inhibitor resistance or intolerance4,43–45. Other therapies have also been designed to improve the cytopenia and to target the marrow fibrosis. Selected ongoing clinical trials are shown in Table 1 A and B.
Table 1. Selected clinical trials.
A) Ongoing clinical trials; recruiting patients
drug | mechanism of action | ongoing clinical trial recruiting | MF patients |
---|---|---|---|
Pacritinib | jak inhibitor |
phase 3 : Pacritinib 200mg BID vs P/C therapy (PACIFICIA) |
≥ int1 risk platelets < 50,000/µL JAK inh naive (1st L) limited exposure JAK inh (2nd L) |
Pegasys | interferon | phase 1/2 : Pegasys + ruxolitinib (RUXOPeg) phase2/3 : COMBI-I and COMBI-II |
≥ int1 risk JAK inh naive (1st L) |
Rogepinterferon alpha 2b |
interferon | phase 2 | prePMF/low/int1 risk |
Imetelstat | telomerase inhibitor | phase 3 : Imetelstat vs BAT | int2/high risk R/R JAK inh (2nd L) |
Pelabrasib | BET inhibitor |
phase 3 : Pelabrasib vs placebo + ruxolitinib (MANIFEST-2) |
≥ int1 risk JAK inh naive (1st L) |
BMS-986158 | BET inhibitor | phase 1b/2 : monotherapy or + ruxolitinib or + fedratinib |
≥ int1 risk JAK inh naive (1st L) |
ABBV-744 | BET inhibitor | phase 1b : monotherapy or + navitoclax or + ruxolitinib |
int2/high risk R/R /intolerant JAK inh (2nd L) |
Bromedemstat | LSD1 inhibitor | phase 2 : Bromedemstat + ruxolitinib |
≥ int1 risk A : R/R/intolerant JAK inh (2nd L) B : JAK inh naive (1st L) |
Parsaclisib | PI3Kẟ inhibitor |
phase 3 : Parsaclisib vs placebo + ruxolitinib (LIMBER-313) |
≥ int1 risk JAK inh naive (1st L) |
AUY922 | HSP90 inhibitor | phase 2 | ineligible JAK inh (1st L) R/R JAK inh (2nd L) |
TP-3654 | PIM inhibitor | phase 1/2 | ≥ int1 risk R/R JAK inh (2nd L) ineligible JAK inh (1st L) |
abemaciclib | CDK4/6 inhibitor | phase 1 | ≥ int1 risk inadequate resp JAK inh (2nd L) |
TL-895 | BTK tyrosine kinase inhibitor |
phase 2 Phase 1b/2 : TL-895 + ruxolitinib |
≥ int1 risk R/R/intolerant JAK inh (2nd L) ineligible JAK inh (1st L) JAK inh naive (1st L) suboptimal resp JAK inh (2nd L) |
Navitoclax | BCL2, BCL-xL,BCL-W inhibitor |
phase 3 : navitoclax + ruxolitinib vs BAT (TRANSFORM-2) |
int2/high risk R/R /intolerant/suboptimal resp JAK inh (2nd L) |
KRT-232 | MDM2 inhibitor |
phase 2/3 : KRT-232 vs BAT phase 1b/2 : KRT-232 + ruxolitinib |
≥ int1 risk failure of JAK inh (2nd L) ≥ int1 risk suboptimal resp JAK inh (2nd L) |
Selinexor | XPO1 inhibitor | phase 2 : selinexor vs P/C therapy phase 2 : selinexor single arm phase 1/2 : selinexor + ruxolitinib |
≥ int1 risk R/R/intolerant JAK inh (2nd L) ≥ int1 risk – JAK inh naive (1st L) |
GB2064 | LOXL2 inhibitor | phase 2a | int2/high risk R/R/intolerant JAK inh (2nd L) ineligible JAK inh (1st L) |
PXS-5505 | LOX inhibitor | phase 1/2a | int2/high risk R/R/intolerant JAK inh (2nd L) ineligible JAK inh (1st L) |
Luspatercept | ActRIIA ligand trapping |
phase 3 : luspatercept vs placebo (INDEPENDENCE) |
anemia on JAK inh |
KER-050 | modified ActRIIA ligand trapping |
phase 2 : monotherapy or + ruxolitinib |
anemia on JAK inh/ineligible JAK inh |
Nivolumab | PD-L1 inhibitor | phase 2 : nivolumab + fedratinib | int2/high risk R/R/suboptimal resp JAK inh (2nd L) |
Tagraxofusp | IL3 fused to diphteria targeting CD123 + LSC |
phase 1 | post-transplant maintenance |
B) Ongoing clinical trials; active but not recruiting patients
drug | mechanism of action | ongoing clinical trial active not recruiting |
MF patients |
---|---|---|---|
Mivebresib | BET inhibitor | Phase 1b : monotherapy or + ruxolitinib or + navitoclax |
≥ int1 risk R/R/intolerant JAK inh (2nd L) |
Navitoclax | BCL2, BCL-xL,BCL-W inhibitor |
phase 3 : navitoclax + ruxolitinib vs ruxolitinib (TRANSFORM-1) |
int2/high risk JAK inh naive (1st L) |
Navitoclax | BCL2, BCL-xL,BCL-W inhibitor |
phase 2 : monotherapy or + ruxolitinib (REFINE) |
int2/high risk R/R/intolerant JAK inh (2nd L) |
Siremadlin Crizanlizumab Sabatolimab Rineterkib NIS793 |
MDM2 inhibitor Monoclonal antibody P-selectin Monoclonal antibody TIM-3 ERK inhibitor Monoclonal antiboby TGFβ1-2 |
phase 1b/2 : Platform Study of Novel Ruxolitinib Combinations in Myelofibrosis Patients (ADORE) |
≥ int1 risk treated with ruxolitinib since 12 weeks (2nd L) |
Abbreviations : BID: twice a day; P/C: per physician choice; Int1: intermediate-1 risk following the DIPSS score; 1st L: first line; JAK inh: JAK inhibitor; Int2: intermediate-2 risk following the DIPSS score; high risk: high risk following the DIPSS score; prePMF: prefibrotic primary myelofibrosis; 2nd L: second line; vs: versus; BAT: Best available therapy; R/R relapsed/refractory; resp: response.
JAK2 inhibitors
Ruxolitinib was the first JAK inhibitor to be approved in the treatment of intermediate and high-risk MF. Ruxolitinib is a JAK1/2 inhibitor without selective effect on JAK2V617F or on oncogenic activation of JAK2WT by CALR or MPL mutants (see Figure 1A). It has become the reference for treatment of MF by reducing general symptoms, splenomegaly and improving the quality of life in around 50% of cases. The main toxicities are anemia and thrombocytopenia, in line with on-target effects. Despite an initial response, around 50% of the patients will discontinue ruxolitinib within 3 years mainly due to a lack/loss of response, cytopenia and/or progression to blast phase. However, it may also exert non-hematological side effects, the most prevalent being weight gain with increased levels of cholesterol and triglycerides and infections such as urinary infections and reactivation of herpes Zoster, tuberculosis and hepatitis B. There is some suspicion of an increased frequency of non-melanoma-skin cancers and B cell non-Hodgkin lymphoma. An abrupt termination may lead to the ruxolitinib discontinuation syndrome with a rapid increase in the spleen volume and a cytokine release syndrome46. Overall, in most patients, ruxolitinib has no significant effect on the progression of the disease and minor effect on the clonal disorder4. Some mutations such as ASXL1 are predictive of a poor response to ruxolitinib. ASXL1, as well as EZH2 and RAS, mutations can be acquired during treatment47. Ruxolitinib has a moderate effect on patient survival, except in patients with a prolonged response to treatment. For these reasons, other JAK2 inhibitors have been developed.
Figure 1. Targets and pathways of new therapies alone or in combination with JAK2 inhibitor (derived from reference 43).
A) Signaling pathways, B) Apoptosis, C) Cell cycle, D) Epigenetic regulation, E) Myelofibrosis. The Figures have been created with bioRender.
Fedratinib, a JAK2/FLT3 inhibitor, is the second approved JAK inhibitor in MF. Results are similar to ruxolitinib, with better splenomegaly response, but an increased non-hematological toxicity48. Fedratinib is indicated for patients intolerant or resistant to ruxolitinib, although in the US it can be used as a frontline therapy. Fedratinib has an increased gastrointestinal toxicity in comparison to ruxolitinib. There may be an increased risk of Wernicke encephalopathy due to thiamine deficiency as a consequence of a gastrointestinal toxicity and a controversial inhibition of the thiamine transporter-2. It is recommended to follow the thiamine levels during therapy49.
Pacritinib is a JAK2/FLT3 inhibitor and also inhibits IRAK1, implicated in myddosome, which regulates the synthesis of numerous pro-inflammatory cytokines including type I IFN. S100A8/9 (calprotectin) and IL33 that induce myddosome signaling are involved at the pre-fibrosis stage by reprogramming MSC into myofibroblasts22,50. However, no clear evidence exists that pacritinib inhibits marrow fibrosis development. Pacritinib also targets the activin receptor-type 1 (ACVR1/ALK2). The best indication of pacritinib concerns patients with marked thrombocytopenia and/or anemia. The main specific side effect is gastrointestinal toxicity, as described for fedratinib. In addition, in the initial clinical trial, it was suggested that pacritinib was associated with an increased risk of severe bleeding and cardiac events. It was not confirmed in the recent clinical trials51.
Momelotinib, recently FDA approved, is a JAK1/2 inhibitor that also targets ACVR1/ALK2 that transduces the BMP signal and regulates the liver synthesis of hepcidin43,52. Hepcidin is involved in the anemia of inflammatory disorders by sequestering the iron. Therefore, the principal advantage of momelotinib is to alleviate anemia4. The main specific side effects are thrombocytopenia, gastrointestinal toxicity, headache, peripheral sensory neuropathy and first-dose effect (dizziness, hypotension, or flushing).
In clinical trials, momelotinib and pacritinib exhibit similar effects as ruxolitinib on general symptoms and splenomegaly, but may correct the anemia in patients, alleviating the need for RBC transfusion4. Pacritinib seems to be a safe option for MF patients with severe thrombocytopenia53. It has to be confirmed by the undergoing randomized phase III study (PACIFICA).
None of these inhibitors display a selective inhibition on the oncogenic activation of JAK2, thus they do not induce molecular remission, whereas a strong JAK2 inhibition induces profound cytopenia. In agreement, resistance is not associated with JAK2 mutations in patients. When mutations in the RAS pathway occur during therapy, they can be present either in the JAK2V617F clone or JAK2WT cells54.
Therefore, there is a need for the development of JAK2V617F selective inhibitors or molecules targeting the activation of JAK2WT by CALR and MPL mutants (see last section).
IFNα
IFNα is an old therapy of MPN, as it has been used since 1988 in ET and PV55,56. IFNα has become one of the major therapies in ET and PV for two reasons: (a) the toxicity of IFNα has been alleviated by using prolonged half-life type 1 IFN, and (b) it is the only available treatment that can act on the clonal disorder and may induce a deep molecular remission (in 25% of the cases)57. The efficacy of IFNα on the clonal disorder is dependent on the dose and the type of disease driver mutation. It is decreasingly efficient in targeting homozygous JAK2V617F, heterozygous JAK2V617F, CALR type-2 and CALR type-158. However, the hematological remission is not dependent on the disease driver mutation. The role of associated mutations in the resistance to IFNα therapy is controversial. It clearly selects DNMT3A mutations whether they are biclonal or associated with JAK2V617F59. The mechanism by which IFNα therapy acts on the clonal disorder is not completely elucidated. There is strong evidence using mouse models that it targets JAK2V617F HSCs by inducing: (a) their entry in the cell cycle as normal HSCs, but without reentry in quiescence, thus principally leading to exhaustion of mutated cells (b) their apoptosis and eventually senescence through ROS accumulation and p53 induction, and (c) a shift of JAK2V617F HSCs to MK/myeloid biased HSCs with lower long-term reconstitution capacities57,60,61. (Figure 2A)
Figure 2. Approaches selectively targeting the disease drivers.
A) Interferon alpha, B) JAK2V617F, C) Calreticulin mutants. The Figures have been created with bioRender.
It is suggested that IFNα targets JAK2V617F HSCs, and to a much lower extent CALRmut HSCs, because JAK2V617F primes HSCs to an IFNα response by increasing STAT1 levels and activation, and consequently inducing expression of interferon stimulated genes (ISG)62. The effects of IFNα appear dependent of p53 and, although not demonstrated clinically, one has to exert caution with respect to the use of IFNα therapy in the presence of a TP53 mutation. In addition, IFNα may induce an immune response against the malignant cells. Finally, it has been shown that the induction of the p38 pathway by IFNα through PKCδ and ULK1 is involved in the therapeutic effect of IFNα63.
At present, the use of an IFNα therapy in MF is limited by its toxicity with, as seen in its first trials, a low effect on the splenomegaly. However, preliminary results suggest that pegylated IFN may be efficient in MF, even in high-risk MF. As in PV, the treatment may decrease the JAK2V617F VAF and seems to have an effect on the survival in responder patients. It remains that treatment was stopped in nearly 75% of the patients due either to disease progression or intolerance64. Thus, it will be important to improve the IFN therapy to better target LSCs and to limit treatment length.
Combination of IFNα with ruxolitinib is being tested in clinical assays, although ruxolitinib as a JAK1/2 inhibitor decreases IFNα signaling65. However, ruxolitinib by its powerful anti-inflammatory effect may increase the tolerance to IFNα. Preliminary results suggest some efficacy of this combination in certain PV and MF patients, even those previously intolerant to IFNα therapy, with a decrease in JAK2V617F VAF and in fibrosis65,66. A recent preclinical study in Jak2V617F knock-in mice suggested that the use of fedratinib, a JAK2 inhibitor seems more efficient and may allow a decrease in the IFNα dose, and thus to have less toxic effects67.
Four other combinations have been experimentally efficient in preclinical studies (see Figure 2A):
Arsenic trioxide with IFNα more efficiently targets the JAK2V617F LSC than IFNα alone, allowing long-term remission68. The combination increases PML overexpression and enhances PML nuclear bodies activity through sumoylation/oligomerization, leading to senescence68.
A ROCK inhibitor (fasudil) enhanced the effects of IFNα on JAK2V617F hematopoietic cells in a mouse model63.Activation of ROCK1/2 acts as a feedback regulator of IFNα signaling by interacting with UKL1.
Induction of p53 plays an important role in targeting JAK2V617F clone by IFNα. Combination of IFNα with a MDM2 inhibitor decreased the transplantability of human JAK2V617F HSPC into immunodeficient mice69. However, no clear synergy between IFNα and a MDM2 inhibitor (idasanutlin) was found in a phase 1 clinical trial in PV70.
Combination of IFNα and 5-azacytidine seems extremely efficient in preclinical models71.
Overall, the risk of all these combinations is to increase the hematological toxicity of IFNα.
Molecules in clinical development
Telomerase inhibitor (Figure 1D)
Both telomere shortening and reactivation were observed in MPN. A molecule called imetelstat is a 13-mer lipid-conjugated oligonucleotide that inhibits telomerase activity. After initial testing in ET, imetelstat was tested in MF with a moderate effect on symptoms and spleen volume reduction, but apparently increased survival43,72. A phase 3 clinical trial is ongoing in MF with the goal to demonstrate an effect on overall survival73. Liver toxicity, although transient, is a limitation, as well as cytopenias. Thus, imetelstat could be an alternative for patients that are intolerant or resistant to JAK inhibitors and could represent one of the rare treatments that may induce disease modification.
Most other treatments are developed, essentially, in combination with ruxolitinib.
Epigenetic regulator inhibitors (Figure 1D)
BET inhibitors
The Bromodomain and Extra-Terminal motif (BET) proteins interact with acetylated histones and transcription factors to induce gene expression such as MYC and genes downstream, NF-κB and TGF-β. Either small molecule inhibitors or BET degraders have been developed. In mouse models, the association of JQ-1 BET inhibitor and ruxolitinib was synergic in reducing fibrosis and the clonal disorder74. The effects are due to inhibiting NFκB transactivation and thus inflammation. In addition, a BET degrader targets the LSC in xenografts75.
Clinical trials are on-going using another BET inhibitor (pelabresib) alone or associated with ruxolitinib. Preliminary results suggest an efficiency with a decrease in JAK2V617F VAF, an effect on the marrow fibrosis and on anemia76.
LSD1 inhibitor
LSD1 (KDM1A), an H3K4 demethylase, is involved in transcriptional repression by the coREST complex77. It associates with GFI1/GFI1b and plays an important role in LSC self-renewal and differentiation. In addition, it regulates STAT3 activity. In JAK2V617F mouse models, LSD1 inhibition alleviates the myeloproliferative disorder and reduces MF acting in synergy with ruxolitinib78. It reduces the inflammation by decreasing inflammatory cytokine synthesis including IL8.
Bomedemstat, a LSD1 inhibitor, has been tested in human with some effects on the spleen volume, general symptoms, and the VAF of some mutations, such as ASXL1 and fibrosis77.
Protein arginine N-methyltransferase 5 (PRMT5)
PRMT5 is a protein arginine-methylase, which methylates histone and non-histone proteins. PRMT5 is phosphorylated by JAK2V617F, impairing its methylase activity, which leads to HSC expansion and to increased erythroid differentiation79. However, it was subsequently shown that in MPN, PRMT5 is overexpressed, which positively regulates E2F1 target genes. In MPN mouse models, a PRMT5 inhibitor (C220) decreases myeloproliferation and systemic inflammation. Combination with ruxolitinib was more effective than individual therapy80. The PRMT5 inhibitor, PRT543, is now being evaluated in MF patients44.
HDAC inhibitors (Figure 1A and D)
HDACs are a large family of proteins that can deacetylate histones leading to gene repression but also gene activation. They also deacetylate non-histone proteins such as HSP90. The rationale to develop HDAC inhibitors was that HDACs 1-3 play an important role in HSC biology and self-renewal and HDAC1/2 are involved in erythroid and MK differentiation81. In addition, HSP90 behaves as chaperone for JAK2V617F, their interaction requiring HSP90 deacetylation by HDAC682. In vitro combinations of a pan-HDAC inhibitor and ruxolitinib were very effective83. However, clinical trials were quite disappointing in MF with regards to toxicity (neutropenia and thrombocytopenia)84. Specific inhibitors of HSP90/HDAC6 or HDAC11 involved in oncogenic JAK/STAT signaling could be more relevant82,85.
Inhibitors of signaling molecules (Figure 1A)
Numerous signaling pathways are induced by the oncogenic activation of JAK2. Ruxolitinib only partially blocks these signaling pathways, such as the PI3K/AKT/mTOR and ERK/MAPK pathways, especially in CALRmut MPN.
Inhibitors of the PI3K pathway
The PI3K pathway through AKT and mTOR plays an important role in both cell proliferation and survival for key targets p27 and BAD. Both in vitro studies and MPN mouse models have shown that pan-PI3K, mTOR, and AKT inhibitors have major effects on the MPN, with a synergistic effect with ruxolitinib86–88. However, pan-PI3K inhibitors have an important dose-dependent toxicity. PI3Kδ is the most expressed PI3K isoform in MF CD34+ cells. Parsaclisib, a new generation PI3Kδinhibitor, is being tested in phase 3 clinical trials of MF in association with ruxolitinib4,43,60,89. However, the clinical trial has been recently discontinued due to a lack of efficiency on the reduction of spleen volume. Pleckstrin-2 could be another target of this pathway90.
MEK and ERK inhibitors (Figure 1 A and E)
The RAS/MAPK pathway plays an important role in the resistance to ruxolitinib, with the frequent occurrence or selection of mutations on this pathway during ruxolitinib treatment54. Mouse models have shown that MEK/ERK inhibition increases the effects of JAK2 inhibition on the myeloproliferation, MF and systemic inflammation by decreasing the level of numerous pro-inflammatory cytokines and osteopontin33,91. The efficacy of a combination between an ERK inhibitor (rineterkib) and ruxolitinib is being tested in a clinical trial43.
PIM (Figure 1 B and C)
The PIM family is composed of serine-threonine kinases that are the direct transcriptional targets of STAT5. They are constitutively active kinases only regulated by their expression. They cooperate with MYC in oncogenesis. PIM1 is overexpressed in MF CD34+ cells. Genetic deletion or pharmacologic inhibition of PIM together with ruxolitinib has a major effect on the myeloproliferative disorder, reducing the fibrosis by decreasing the TGF-β1 level92–94.
HSP90
HSP90 is as a molecular chaperone with numerous substrate proteins, including JAK2V617F. Its inhibition leads to the degradation of total and phospho-JAK2. In MPN mouse models, the combination of ruxolitinib with an HSP90 inhibitor was more efficient than ruxolitinib alone by further decreasing JAK/STAT signaling95,96. An initial clinical trial with AUY922 induces a severe non-hematologic toxicity97. A combination of PU-H71 with ruxolitinib is currently being evaluated in MF patients.
Apoptosis inducers (Figure 1C) and Cell cycle inhibitors (Figure 1D)
Targeting key molecules implicated in proliferation and cell survival downstream of JAK2 appears as an interesting therapeutic approach.
CDK4/6 inhibitor
CDK4/6 are key molecules in cell cycle entry as they phosphorylate Rb when activated. In contrast to CDK4, CDK6 is involved in stress hematopoiesis and hematological malignancies and regulates transcription in both kinase-dependent and -independent manners. CDK6 is overexpressed in CD34+ cells from MPN. JAK2V617F regulates CDK6 through CDC25A or PIM. Several CDK4/CDK6 inhibitors, including palbociclib, have been approved for the treatment of solid tumors, with an acceptable hematological toxicity. CDK6 ablation has a major effect on the myeloproliferative disorder, but also on inflammation inhibiting NFκB and TGF-β signaling98. CDK4/CDK6 inhibitors exert a synergistic therapeutic effect with ruxolitinib and a PIM inhibitor in mouse models92,99. CDK6 PROTAC is under development100.
Aurora A kinase inhibitor
Aurora A is a serine threonine kinase involved in G2/M transition and the organization of the spindle, regulating many other targets. Aurora A inhibition in acute megakaryoblastic leukemia leads to differentiation101. In mouse models, its inhibition ameliorated MF102. A phase 1 clinical study was performed with a response in 30% of cases, including a decrease in MF and JAK2V617F/CALR VAF without major toxicity103.
BCL2 family inhibitor
The BCL2 family includes pro-survival molecules such as BCL2, BCL-xL and MCL1 and pro-apoptotic molecules. BCL2 and MCL1 regulate the survival of early stages of hematopoiesis whereas BCL-xL is indispensable for erythroblasts and MKs. BCL-xL and BCL2 are direct targets of STAT5 and indirect targets of the PI3K pathway and are overexpressed in MPN60. Venetoclax, a BCL2 inhibitor, is now a promising therapy in AML. In MPN, there is a need to also target BCL-xL. Navitoclax is an inhibitor of BCL2, BCL-xL and BCL-W. In JAK2V617F cell lines, there was a synergy between JAK2 inhibition and navitoclax, also leading to reversal of the ruxolitinib resistance104. Phase 2 clinical trials of this combination were promising with a very significant decrease in the spleen volume, a decrease of fibrosis, including some patients with a resolution of the marrow fibrosis and of the JAK2V617F/CALR VAF44,105. Furthermore, the thrombocytopenia was manageable and, surprisingly, the anemia was partially corrected. Phase 3 clinical trials are ongoing. This combination seems to be one of the most effective.
MDM2 inhibitor and the p53 pathway
MDM2 is an E3 ubiquitin ligase that negatively regulates p53 by degradation. p53 is a potent negative regulator of JAK2V617F signaling that is downregulated by MDM2 overexpression by JAK2V617F106. Results of a phase 2 trial with navtemadlin MDM2 inhibitor were encouraging, and further clinical trials including a phase 3 trial are being conducted in monotherapy or in combination with ruxolitinib in MF107. Some specific inhibitors of PPM1D, another P53 regulator, have been developed that could be combined with MDM2 inhibitors at lower doses to avoid gastro-toxicity108. However, the risk of such treatments is to select TP53 mutated subclone, thus patients with a TP53 mutation must be excluded109.
Selective inhibitors of nuclear export (Figure 1D)
A shRNA screening on a JAK2V617F cell line identified a particular sensitivity to the inhibition of the nuclear export transport (NE) machinery110. These results were confirmed in primary MF CD34+ cells and in a mouse model with a combination of a specific NE compound (selinexor) and ruxolitinib110. The effects of inhibiting NE are presumably related to the accumulation of tumor suppressor proteins in the nucleus, especially p53. A phase 2 clinical trial with selinexor is ongoing in MF JAK2 inhibitor intolerant patients111.
Other inducers of apoptosis
LCL-161 is a second mitochondrial activator of caspases (SMAC) mimetic that antagonizes inhibitors of apoptosis (IAP). In preclinical studies, LCL-161 induced JAK2V617F cell apoptosis that was rescued by JAK2 inhibition112. Thus a phase-2 clinical trial in monotherapy was performed and gave encouraging results in old high-risk patients113.
Targeting the marrow fibrosis (Figure 1E)
These approaches are based on the marrow fibrosis mechanism.
First Approach
The first approach targets the external cues involved in fibrosis development or their signaling.
A first trial was performed using an anti-TGF-β1antibody, but it only includes 3 patients with a response essentially on the anemia scale114. A second assay was conducted with AVID 200, a TGF-β1/3 ligand trap (TGF-β receptor ectodomains fused to a human Fc domain). Treatment of GATA1low mice by AVID200 reduced marrow fibrosis115. A preliminary clinical trial led to the improvement of thrombocytopenia. Another trial is programmed with a new anti-TGF-β1 monoclonal antibody (MoAb).
Approaches blocking inflammatory cytokines or their signaling such as IL8, IL13 or IL1β and S100A8/S100A9 were promising in MPN mouse models with synergistic or additive effects with ruxolitinib28,31,32,116. Interestingly, pacritinib by inhibiting IRAK1 may inhibit the signaling of both IL1β and S100A8/S100A922.
JAK2V617F decreases the number of nestin cells involved in HSC regulation by inducing their apoptosis, due to an IL1β-induced damage of sympathetic nerves that innerve nestin cells117. Mirabegron, a β-3 sympathomimetic agonist, has been tested in a phase 2 clinical trial leading to a slight increase in nestin cells and decrease in fibrosis, but without altering the JAK2V617F VAF118.
Second Approach
Serum amyloid P (pentraxin 2) is capable of suppressing fibrosis of many organs by inhibiting the differentiation of monocytes into fibrocytes and by acting on macrophages and neutrophils. The recombinant form, PRM-151, has been shown to inhibit in vitro fibrocyte differentiation from PMF36. In addition, it prolonged survival of mice xenotransplanted with PMF hematopoietic cells. Therefore, PRMT-151 has been tested in a phase 2 clinical trial with a regression of fibrosis and improvement of cytopenia in some patients119. It has been recently also tested in association with ruxolitinib in phase 1 and 2 clinical trials with an acceptable toxicity120.
Another closely related approach consists of the use of a SLAMF7 antibody to prevent differentiation of monocytes into fibrocytes37.
Third Approach
This approach concerns the targeting of Lysyl oxidase-like-2 (LOXL2) and P-selectin.
LOXL2 is an enzyme that stabilizes the ECM by crosslinking collagen that is also involved in MK expansion induced by PDGFB24. While it is overexpressed by MKs in marrow fibrosis mouse models, its pharmacological inhibition reduces marrow fibrosis121. A phase 2 clinical trial using an anti-LOXL2 MoAb was conducted in MF patients alone or in association with ruxolitinib, the results being disappointing122. A phase 2 clinical trial is ongoing using an inhibitor of LOXL2, GB2064. In 4 patients a reduction in collagen fibrosis was observed at 6 months123.
P-selectin is a glycoprotein that is present in platelet and MK α-granules. It is translocated to the cell surface after activation. P-selectin is involved in the emperipolesis of granulocyte precursors by MKs, leading to TGF-β1 release35. In the GATA1low mouse model, SELP (P-selectin gene) ablation or P-selectin blockage by a MoAb impairs MF development124. A phase 1 trial is programmed using a combination of ruxolitinib and a MoAb44.
Targeting cytopenia
All the previous approaches aim also to correct the cytopenia by modifying the disease. In addition, specific therapies for anemia are being tested in MF beyond the classical therapies (androgens, IMID agents and ESA)4,125. The first consists of a ligand trap strategy using the extracellular domain of ACVRIIB and IIA fused to the human IgG Fc domain for luspatercept and sotatercept, respectively. These ligand traps bind some members of the TGFβ superfamily, such as GDF11 that negatively regulates late stages of erythroid differentiation through SMAD2/3126. In addition, they may decrease hepcidin synthesis by trapping some BMPs involved in its synthesis and by increasing the level of erythroferrone as a consequence of an increased erythroid maturation and thus they mobilize the iron store. Both ligand traps have been tested in phase 2 clinical trials alone or in association with ruxolitinib to correct the anemia of MF. Around one third of the patients had a clinical response43,125. In addition, such therapies may have an effect on cachexia of advanced MF patients.
The other approaches aim to target hepcidin, which is regulated at the transcriptional level by IL6 (and other members of the family) through the GP130/JAK2/1/STAT3 pathway and by BMPs1/6, the first pathway being regulated by inflammation, the second by an iron overload. Momelotinib targets both pathways of hepcidin regulation52. In addition, an ACVR1/ALK2 inhibitor (BMP pathway) is being tested in combination with ruxolitinib in anemia of MF43,125.
In the future, other approaches could be used in MF, including hepcidin antagonists, whereas hepcidin mimetics appear quite efficient in the therapy of PV erythrocytosis.
New approaches specifically targeting the clonal disease
Presently, most new therapies are based on the association of ruxolitinib with other molecules. However, the risk of drug combination is increased toxicity. It is important to develop new approaches targeting either specifically the disease driver mutants (JAK2V617F, CALR or MPL mutants) or directly the LSC.
These last years, there has been some progress in the development of an immunotherapy against CALRmut and JAK2V617F MPNs.
Immunotherapy
JAK2V617F and CALRmut MPN clones may escape T cell surveillance, even if CD4 or CD8 T cells directed against these mutants exist. These immune cells are rendered non-functional by the PD1/PD-L1 axis. However, a phase 2 clinical trial using pembrolizumab, an anti-PD1 antibody, failed to induce a clinical response127.
CALRmut generate a new C-terminus, with neo-antigens that could be targeted by immunotherapy. However, this approach may be limited by several factors: (a) MHC-1 having a high affinity for these neo-epitopes is under-represented in MPN patients128; (b) huge levels of soluble mutated CALR are present that inhibit the phagocytosis of dying cancer cells by dendritic cells and suppress the effects of PD-1 blockade129; and (c) CALR is implicated in the peptide loading on MHC-1130. A first trial using a peptide vaccine showed no clinical response, although a strong immune response was observed mainly involving CD4+ T cells131. Trials are ongoing using a vaccine associated with an immune checkpoint inhibitor. Alternatively, it has been shown that a heteroclitic peptide with an optimized presentation by MHC-1 may permit to overcome this immunosuppressive state128.
LSC targeting
In MF, LSCs are skewed towards the MK lineage and aberrantly express MK/platelet antigens, such as G6B, defining new targets for immunotherapy132. In addition, CD123, the IL3α-receptor, is expressed on LSCs of numerous myeloid malignancies. Presently, tagraxofusp, a fusion protein consisting of IL3 fused to diphteria toxin has been tested in a phase 1/2 clinical trial, but with modest results133.
Direct targeting of the mutated disease drivers (Figure 2B and C)
Unexpectedly, CALRmut can be targeted more easily than JAK2V617F.
Using a large screening approach, it has been shown that inhibition of N-glycosylation by several molecules can target the oncogenic CALRmut signaling by inhibiting the MPL membrane expression134. Such an approach could be extended to MPL mutants. In addition, CALRmut multimerization is dependent on zinc, thus zinc chelators could be a valuable approach135.
The development of MoAbs targeting specifically the CALRmut induced positive expectations. Anti-CALRmut antibodies may either impair abnormal signaling of MPL/JAK2 complexes or induce an immune reaction. Three different anti-CALRmut MoAbs, namely B3, 4D7 and INCA033989, were generated136–138. 4D7 and INCA033989 disrupt CALR mutant/MPL signaling136,137. These antibodies inhibit in vitro the growth of CALRmut/MPL cell lines and patient cells including in xenografts. In addition, this approach appears to have a major effect on the disease development in a murine model by targeting the LSC137,138. A clinical trial is in progress with INCA033989. Another antibody against CALRmut injected in CALRdel52 mice model led to a very rapid normalization of platelet count and a decrease of LSK139. This response may reflect immunodepletion rather than signaling interruption. Finally, a synthetic peptide inhibiting the interaction between MPL and CALRmut was able to inhibit the constitutive MPL/JAK2 signaling in vitro140.
Concerning JAK2, due to the frequently acquired resistance to type I JAK inhibitors (ruxolitinib, fedratinib), two type II JAK inhibitors that interact with a JAK2-inactive conformation were efficiently tested in preclinical studies, but were toxic141. A new type II JAK2 inhibitor (AJ1-10502) showed improved efficacy in comparison to ruxolitinib, with a selective effect on JAK2V617F cells142.
Specific JAK2V617F allosteric inhibitors theoretically seem the more straightforward approach to directly impact the clone without inducing cytopenia. Numerous progresses have been obtained in the structure of JAK2V617F by mutational approaches and by ultrastructural analysis143–145. Recently, a cryo-EM structure of the complex between IFNAR2/JAK1V657F, the homologous mutation of JAK2V617F, has been obtained146. Despite the limitation using JAK1, this new structure indicated dimerization of the mutated PK domains of JAK1. New small molecules inhibiting the dimerization of JAK2V617F may be promising. A limitation of this approach is that the conformation of JAK2V617F is close to the conformation of JAK2WT associated with the IFNγ receptor after ligand binding, raising the risk that such molecules may thus inhibit IFNγ signaling144.
Another approach will be to target JAK2V617F degradation. This has been recently reported in ALL by determining the structure of the interaction of ruxolitinib and baricitinib with JAK2147. A better knowledge of the JAK2V617F structure may also allow the development of specific PROTAC.
A last approach is to target the conformational differences between interaction of JAK2V617F or JAK2WT interacting with MPL and EPOR. This strategy using diabodies has been successful in targeting the EPOR/JAK2V617F interaction148.
In conclusion
Presently, efforts are geared towards finding new therapies that would profoundly modify the disease either based on drug combination or on the development of new compounds, some directly targeting the disease drivers. All these efforts would likely lead in the future to major advances in the treatment of MF.
The peer reviewers who approve this article are:
Douglas A Tremblay, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
Alessandro Maria Vannucchi, Department of Hematology, University of Florence, Florence, Italy
Brady Stein, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
Funding Statement
Funding to SNC is acknowledged from Ludwig Institute for Cancer Research, Fondation contre le cancer, Salus Sanguinis and Fondation “Les avions de Sébastien”, projets Action de recherché concertée (ARC) 16/21-073, Projet de recherche FNRS n°T.0043.21 and WelBio F 44/8/5 - MCF/UIG – 10955.
Funding to IP, CM and WV is acknowledged from la Ligue Nationale contre le Cancer (équipe labélisée), INCA-Plbio 2021 and Grex (INCA).
Funding to VH is acknowledged from FNRS (Clinical Researcher), Salus Sanguinis, Fondation Roi Baudouin, Fonds Joseph Maisin and Fondation Saint-Luc.
The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
References
- 1. Luque Paz D, Kralovics R, Skoda RC: Genetic basis and molecular profiling in myeloproliferative neoplasms. Blood. 2023; 141(16): 1909–1921. 10.1182/blood.2022017578 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 2. Vainchenker W, Kralovics R: Genetic basis and molecular pathophysiology of classical myeloproliferative neoplasms. Blood. 2017; 129(6): 667–79. 10.1182/blood-2016-10-695940 [DOI] [PubMed] [Google Scholar]
- 3. Ortmann CA, Kent DG, Nangalia J, et al. : Effect of mutation order on myeloproliferative neoplasms. N Engl J Med. 2015; 372(7): 601–12. 10.1056/NEJMoa1412098 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 4. Tefferi A: Primary myelofibrosis: 2023 update on diagnosis, risk-stratification, and management. Am J Hematol. 2023; 98(5): 801–821. 10.1002/ajh.26857 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 5. Tefferi A, Gangat N, Pardanani A, et al. : Myelofibrosis: Genetic Characteristics and the Emerging Therapeutic Landscape. Cancer Res. 2022; 82(5): 749–63. 10.1158/0008-5472.CAN-21-2930 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Barbui T, Thiele J, Vannucchi AM, et al. : Rationale for revision and proposed changes of the WHO diagnostic criteria for polycythemia vera, essential thrombocythemia and primary myelofibrosis. Blood Cancer J. 2015; 5(8): e337. 10.1038/bcj.2015.64 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Constantinescu SN, Vainchenker W, Levy G, et al. : Functional Consequences of Mutations in Myeloproliferative Neoplasms. HemaSphere. 2021; 5(6): e578. 10.1097/HS9.0000000000000578 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Williams N, Lee J, Mitchell E, et al. : Life histories of myeloproliferative neoplasms inferred from phylogenies. Nature. 2022; 602(7895): 162–8. 10.1038/s41586-021-04312-6 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 9. How J, Hobbs GS, Mullally A: Mutant calreticulin in myeloproliferative neoplasms. Blood. 2019; 134(25): 2242–8. 10.1182/blood.2019000622 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 10. Elf S, Abdelfattah NS, Chen E, et al. : Mutant Calreticulin Requires Both Its Mutant C-terminus and the Thrombopoietin Receptor for Oncogenic Transformation. Cancer Discov. 2016; 6(4): 368–81. 10.1158/2159-8290.CD-15-1434 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Chachoua I, Pecquet C, El-Khoury M, et al. : Thrombopoietin receptor activation by myeloproliferative neoplasm associated calreticulin mutants. Blood. 2016; 127(10): 1325–35. 10.1182/blood-2015-11-681932 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 12. Araki M, Yang Y, Masubuchi N, et al. : Activation of the thrombopoietin receptor by mutant calreticulin in CALR-mutant myeloproliferative neoplasms. Blood. 2016; 127(10): 1307–16. 10.1182/blood-2015-09-671172 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 13. Pecquet C, Chachoua I, Roy A, et al. : Calreticulin mutants as oncogenic rogue chaperones for TpoR and traffic-defective pathogenic TpoR mutants. Blood. 2019; 133(25): 2669–81. 10.1182/blood-2018-09-874578 [DOI] [PubMed] [Google Scholar]
- 14. Masubuchi N, Araki M, Yang Y, et al. : Mutant calreticulin interacts with MPL in the secretion pathway for activation on the cell surface. Leukemia. 2020; 34(2): 499–509. 10.1038/s41375-019-0564-z [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 15. Pecquet C, Papadopoulos N, Balligand T, et al. : Secreted mutant calreticulins as rogue cytokines in myeloproliferative neoplasms. Blood. 2023; 141(8): 917–29. 10.1182/blood.2022016846 [DOI] [PubMed] [Google Scholar]
- 16. Di Buduo CA, Abbonante V, Marty C, et al. : Defective interaction of mutant calreticulin and SOCE in megakaryocytes from patients with myeloproliferative neoplasms. Blood. 2020; 135(2): 133–44. 10.1182/blood.2019001103 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Jutzi JS, Marneth AE, Jiménez-Santos MJ, et al. : CALR-mutated cells are vulnerable to combined inhibition of the proteasome and the endoplasmic reticulum stress response. Leukemia. 2023; 37(2): 359–69. 10.1038/s41375-022-01781-0 [DOI] [PubMed] [Google Scholar]
- 18. Hermange G, Rakotonirainy A, Bentriou M, et al. : Inferring the initiation and development of myeloproliferative neoplasms. Proc Natl Acad Sci U S A. 2022; 119(37): e2120374119. 10.1073/pnas.2120374119 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Sousos N, Ní Leathlobhair M, Simoglou Karali C, et al. : In utero origin of myelofibrosis presenting in adult monozygotic twins. Nat Med. 2022; 28(6): 1207–11. 10.1038/s41591-022-01793-4 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 20. Vainchenker W, Plo I, Marty C, et al. : The role of the thrombopoietin receptor MPL in myeloproliferative neoplasms: recent findings and potential therapeutic applications. Expert Rev Hematol. 2019; 12(6): 437–48. 10.1080/17474086.2019.1617129 [DOI] [PubMed] [Google Scholar]
- 21. Grinfeld J, Nangalia J, Baxter EJ, et al. : Classification and Personalized Prognosis in Myeloproliferative Neoplasms. N Engl J Med. 2018; 379(15): 1416–30. 10.1056/NEJMoa1716614 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 22. Mascarenhas J, Gleitz HFE, Chifotides HT, et al. : Biological drivers of clinical phenotype in myelofibrosis. Leukemia. 2023; 37(2): 255–64. 10.1038/s41375-022-01767-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Malara A, Gruppi C, Abbonante V, et al. : EDA fibronectin-TLR4 axis sustains megakaryocyte expansion and inflammation in bone marrow fibrosis. J Exp Med. 2019; 216(3): 587–604. 10.1084/jem.20181074 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 24. Eliades A, Papadantonakis N, Bhupatiraju A, et al. : Control of megakaryocyte expansion and bone marrow fibrosis by lysyl oxidase. J Biol Chem. 2011; 286(31): 27630–8. 10.1074/jbc.M111.243113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Budi EH, Schaub JR, Decaris M, et al. : TGF-β as a driver of fibrosis: physiological roles and therapeutic opportunities. J Pathol. 2021; 254(4): 358–73. 10.1002/path.5680 [DOI] [PubMed] [Google Scholar]
- 26. Chagraoui H, Komura E, Tulliez M, et al. : Prominent role of TGF-beta 1 in thrombopoietin-induced myelofibrosis in mice. Blood. 2002; 100(10): 3495–503. 10.1182/blood-2002-04-1133 [DOI] [PubMed] [Google Scholar]
- 27. Lecomte S, Devreux J, de Streel G, et al. : Therapeutic activity of GARP:TGF-β1 blockade in murine primary myelofibrosis. Blood. 2023; 141(5): 490–502. 10.1182/blood.2022017097 [DOI] [PubMed] [Google Scholar]
- 28. Melo-Cardenas J, Bezavada L, Crawford JC, et al. : IL-13/IL-4 signaling contributes to fibrotic progression of the myeloproliferative neoplasms. Blood. 2022; 140(26): 2805–17. 10.1182/blood.2022017326 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Kleppe M, Kwak M, Koppikar P, et al. : JAK-STAT pathway activation in malignant and nonmalignant cells contributes to MPN pathogenesis and therapeutic response. Cancer Discov. 2015; 5(3): 316–31. 10.1158/2159-8290.CD-14-0736 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 30. Gleitz HFE, Benabid A, Schneider RK: Still a burning question: the interplay between inflammation and fibrosis in myeloproliferative neoplasms. Curr Opin Hematol. 2021; 28(5): 364–71. 10.1097/MOH.0000000000000669 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Dunbar AJ, Kim D, Lu M, et al. : CXCL8/CXCR2 signaling mediates bone marrow fibrosis and is a therapeutic target in myelofibrosis. Blood. 2023; 141(20): 2508–2519. 10.1182/blood.2022015418 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Leimkühler NB, Gleitz HFE, Ronghui L, et al. : Heterogeneous bone-marrow stromal progenitors drive myelofibrosis via a druggable alarmin axis. Cell stem cell. 2021; 28(4): 637–652.e8. 10.1016/j.stem.2020.11.004 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 33. Brkic S, Stivala S, Santopolo A, et al. : Dual targeting of JAK2 and ERK interferes with the myeloproliferative neoplasm clone and enhances therapeutic efficacy. Leukemia. 2021; 35(10): 2875–84. 10.1038/s41375-021-01391-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Varricchio L, Hoffman R: Megakaryocytes Are Regulators of the Tumor Microenvironment and Malignant Hematopoietic Progenitor Cells in Myelofibrosis. Front Oncol. 2022; 12: 906698. 10.3389/fonc.2022.906698 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Centurione L, Di Baldassarre A, Zingariello M, et al. : Increased and pathologic emperipolesis of neutrophils within megakaryocytes associated with marrow fibrosis in GATA-1(low) mice. Blood. 2004; 104(12): 3573–80. 10.1182/blood-2004-01-0193 [DOI] [PubMed] [Google Scholar]
- 36. Verstovsek S, Manshouri T, Pilling D, et al. : Role of neoplastic monocyte-derived fibrocytes in primary myelofibrosis. J Exp Med. 2016; 213(9): 1723–40. 10.1084/jem.20160283 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 37. Maekawa T, Kato S, Kawamura T, et al. : Increased SLAMF7high monocytes in myelofibrosis patients harboring JAK2 V617F provide a therapeutic target of elotuzumab. Blood. 2019; 134(10): 814–25. 10.1182/blood.2019000051 [DOI] [PubMed] [Google Scholar]
- 38. Villeval JL, Cohen-Solal K, Tulliez M, et al. : High thrombopoietin production by hematopoietic cells induces a fatal myeloproliferative syndrome in mice. Blood. 1997; 90(11): 4369–83. 10.1182/blood.V90.11.4369 [DOI] [PubMed] [Google Scholar]
- 39. Ghanima W, Cooper N, Rodeghiero F, et al. : Thrombopoietin receptor agonists: ten years later. Haematologica. 2019; 104(6): 1112–23. 10.3324/haematol.2018.212845 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Adriaanse FRS, Kamens JL, Vogel P, et al. : A CRISPR/Cas9 engineered MplS504N mouse model recapitulates human myelofibrosis. Leukemia. 2022; 36(10): 2535–8. 10.1038/s41375-022-01684-0 [DOI] [PubMed] [Google Scholar]
- 41. Pikman Y, Lee BH, Mercher T, et al. : MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia. PLoS Med. 2006; 3(7): e270. 10.1371/journal.pmed.0030270 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 42. Dunbar A, Bowman RL, Park Y, et al. : JAK2V617F reversible action shows an essential requirement for JAK2V617F in Myeloproliferative Neoplasms. bioRxiv. 2022. 10.1101/2022.05.18.492332 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Chifotides HT, Bose P, Masarova L, et al. : SOHO State of the Art Updates and Next Questions: Novel Therapies in Development for Myelofibrosis. Clin Lymphoma Myeloma Leuk. 2022; 22(4): 210–23. 10.1016/j.clml.2021.10.002 [DOI] [PubMed] [Google Scholar]
- 44. Chifotides HT, Masarova L, Verstovsek S: SOHO State of the Art Updates and Next Questions: Novel Therapeutic Strategies in Development for Myelofibrosis. Clin Lymphoma Myeloma Leuk. 2023; 23(4): 219–231. 10.1016/j.clml.2022.12.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Waksal JA, Harrison CN, Mascarenhas JO: Novel therapeutics and targets in myelofibrosis. Leuk Lymphoma. 2022; 63(5): 1020–33. 10.1080/10428194.2021.2010068 [DOI] [PubMed] [Google Scholar]
- 46. Devos T, Selleslag D, Granacher N, et al. : Updated recommendations on the use of ruxolitinib for the treatment of myelofibrosis. Hematology. 2022; 27(1): 23–31. 10.1080/16078454.2021.2009645 [DOI] [PubMed] [Google Scholar]
- 47. Pacilli A, Rotunno G, Mannarelli C, et al. : Mutation landscape in patients with myelofibrosis receiving ruxolitinib or hydroxyurea. Blood Cancer J. 2018; 8(12): 122. 10.1038/s41408-018-0152-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Saha C, Harrison C: Fedratinib, the first selective JAK2 inhibitor approved for treatment of myelofibrosis - an option beyond ruxolitinib. Expert Rev Hematol. 2022; 15(7): 583–95. 10.1080/17474086.2022.2098105 [DOI] [PubMed] [Google Scholar]
- 49. England JT, Gupta V: Fedratinib: a pharmacotherapeutic option for JAK-inhibitor naïve and exposed patients with myelofibrosis. Expert Opin Pharmacother. 2022; 23(15): 1677–86. 10.1080/14656566.2022.2135989 [DOI] [PubMed] [Google Scholar]
- 50. Venugopal S, Mascarenhas J: The odyssey of pacritinib in myelofibrosis. Blood Adv. 2022; 6(16): 4905–13. 10.1182/bloodadvances.2022007524 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Mascarenhas J: Pacritinib for the treatment of patients with myelofibrosis and thrombocytopenia. Expert Rev Hematol. 2022; 15(8): 671–84. 10.1080/17474086.2022.2112565 [DOI] [PubMed] [Google Scholar]
- 52. Asshoff M, Petzer V, Warr MR, et al. : Momelotinib inhibits ACVR1/ALK2, decreases hepcidin production, and ameliorates anemia of chronic disease in rodents. Blood. 2017; 129(13): 1823–30. 10.1182/blood-2016-09-740092 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 53. Verstovsek S, Mesa R, Talpaz M, et al. : Retrospective analysis of pacritinib in patients with myelofibrosis and severe thrombocytopenia. Haematologica. 2022; 107(7): 1599–607. 10.3324/haematol.2021.279415 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Maslah N, Roux B, Kaci N, et al. : JAK Inhibition Mediates Clonal Selection of RAS Pathway Mutations in Myeloproliferative Neoplasms. Blood. 2022; 140(Supplement 1): 795–6. 10.1182/blood-2022-16771935980680 [DOI] [Google Scholar]
- 55. Bellucci S, Harousseau JL, Brice P, et al. : Treatment of essential thrombocythaemia by alpha 2a interferon. Lancet. 1988; 2(8617): 960–1. 10.1016/s0140-6736(88)92625-6 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 56. Silver RT: Recombinant interferon-alpha for treatment of polycythaemia vera. Lancet. 1988; 2(8607): 403. 10.1016/s0140-6736(88)92881-4 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 57. Kiladjian JJ, Giraudier S, Cassinat B: Interferon-alpha for the therapy of myeloproliferative neoplasms: targeting the malignant clone. Leukemia. 2016; 30(4): 776–81. 10.1038/leu.2015.326 [DOI] [PubMed] [Google Scholar]
- 58. Mosca M, Hermange G, Tisserand A, et al. : Inferring the dynamics of mutated hematopoietic stem and progenitor cells induced by IFNα in myeloproliferative neoplasms. Blood. 2021; 138(22): 2231–43. 10.1182/blood.2021010986 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Knudsen TA, Skov V, Stevenson K, et al. : Genomic profiling of a randomized trial of interferon-α vs hydroxyurea in MPN reveals mutation-specific responses. Blood Adv. 2022; 6(7): 2107–19. 10.1182/bloodadvances.2021004856 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. How J, Garcia JS, Mullally A: Biology and therapeutic targeting of molecular mechanisms in MPNs. Blood. 2023; 141(16): 1922–1933. 10.1182/blood.2022017416 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Rao TN, Hansen N, Stetka J, et al. : JAK2-V617F and interferon-α induce megakaryocyte-biased stem cells characterized by decreased long-term functionality. Blood. 2021; 137(16): 2139–51. 10.1182/blood.2020005563 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Czech J, Cordua S, Weinbergerova B, et al. : JAK2V617F but not CALR mutations confer increased molecular responses to interferon-α via JAK1/STAT1 activation. Leukemia. 2019; 33(4): 995–1010. 10.1038/s41375-018-0295-6 [DOI] [PubMed] [Google Scholar]
- 63. Saleiro D, Wen JQ, Kosciuczuk EM, et al. : Discovery of a signaling feedback circuit that defines interferon responses in myeloproliferative neoplasms. Nat Commun. 2022; 13(1): 1750. 10.1038/s41467-022-29381-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Ianotto JC, Chauveau A, Boyer-Perrard F, et al. : Benefits and pitfalls of pegylated interferon-α2a therapy in patients with myeloproliferative neoplasm-associated myelofibrosis: a French Intergroup of Myeloproliferative neoplasms (FIM) study. Haematologica. 2018; 103(3): 438–46. 10.3324/haematol.2017.181297 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Sørensen AL, Mikkelsen SU, Knudsen TA, et al. : Ruxolitinib and interferon-α2 combination therapy for patients with polycythemia vera or myelofibrosis: a phase II study. Haematologica. 2020; 105(9): 2262–72. 10.3324/haematol.2019.235648 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 66. Kiladjian JJ, Ianotto JC, Soret J, et al. : Final Results of Ruxopeg, a Phase 1/2 Adaptive Randomized Trial of Ruxolitinib (Rux) and Pegylated Interferon Alpha (IFNa) 2a in Patients with Myelofibrosis (MF). Blood. 2022; 140(Supplement 1): 577–78. 10.1182/blood-2022-156389 [DOI] [Google Scholar]
- 67. Ghezzi I, Rodriguez Correa E, Fotopoulou F, et al. : Relevance of dormant cancer stem cells in disease propagation and therapy resistance in JAK2V617driven MPN. HemaSphere. 2023; S208: 387908. [Google Scholar]
- 68. Dagher T, Maslah N, Edmond V, et al. : JAK2V617F myeloproliferative neoplasm eradication by a novel interferon/arsenic therapy involves PML. J Exp Med. 2021; 218(2): e20201268. 10.1084/jem.20201268 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Lu M, Xia L, Li Y, et al. : The orally bioavailable MDM2 antagonist RG7112 and pegylated interferon α 2a target JAK2V617F-positive progenitor and stem cells. Blood. 2014; 124(5): 771–9. 10.1182/blood-2013-11-536854 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Mascarenhas J, Lu M, Kosiorek H, et al. : Oral idasanutlin in patients with polycythemia vera. Blood. 2019; 134(6): 525–33. 10.1182/blood.2018893545 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Lock M, Luque Paz D, Hansen N, et al. : Combination of 5-Azacytidine and Pegifna is able to overcome resistance in JAK2-V617F positive MPN with loss of Dnmt3a. Blood. 2022; 140(Supplement 1): 3876–7. 10.1182/blood-2022-167002 [DOI] [Google Scholar]
- 72. Tefferi A: Telomerase Inhibitor Imetelstat in Essential Thrombocythemia and Myelofibrosis. N Engl J Med. 2015; 373(26): 2580–1. 10.1056/NEJMc1512663 [DOI] [PubMed] [Google Scholar]
- 73. Mascarenhas J, Harrison CN, Kiladjian JJ, et al. : Imetelstat in intermediate-2 or high-risk myelofibrosis refractory to JAK inhibitor: IMpactMF phase III study design. Future Oncol. 2022; 18(22): 2393–402. 10.2217/fon-2022-0235 [DOI] [PubMed] [Google Scholar]
- 74. Kleppe M, Koche R, Zou L, et al. : Dual Targeting of Oncogenic Activation and Inflammatory Signaling Increases Therapeutic Efficacy in Myeloproliferative Neoplasms. Cancer Cell. 2018; 33(1): 29–43.e7. 10.1016/j.ccell.2017.11.009 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 75. Ivanov D, Milosevic Feenstra JD, Sadovnik I, et al. : Phenotypic characterization of disease-initiating stem cells in JAK2- or CALR-mutated myeloproliferative neoplasms. Am J Hematol. 2023; 98(5): 770–783. 10.1002/ajh.26889 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Mascarenhas J, Kremyanskaya M, Patriarca A, et al. : MANIFEST: Pelabresib in Combination With Ruxolitinib for Janus Kinase Inhibitor Treatment-Naïve Myelofibrosis. J Clin Oncol. 2023; Jco2201972. 10.1200/JCO.22.01972 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 77. Gill H: Lysine-Specific Demethylase 1 (LSD1/KDM1A) Inhibition as a Target for Disease Modification in Myelofibrosis. Cells. 2022; 11(13): 2107. 10.3390/cells11132107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Jutzi JS, Kleppe M, Dias J, et al. : LSD1 Inhibition Prolongs Survival in Mouse Models of MPN by Selectively Targeting the Disease Clone. HemaSphere. 2018; 2(3): e54. 10.1097/HS9.0000000000000054 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Liu F, Zhao X, Perna F, et al. : JAK2V617F-mediated phosphorylation of PRMT5 downregulates its methyltransferase activity and promotes myeloproliferation. Cancer cell. 2011; 19(2): 283–94. 10.1016/j.ccr.2010.12.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Pastore F, Bhagwat N, Pastore A, et al. : PRMT5 Inhibition Modulates E2F1 Methylation and Gene-Regulatory Networks Leading to Therapeutic Efficacy in JAK2V617F-Mutant MPN. Cancer Discov. 2020; 10(11): 1742–57. 10.1158/2159-8290.CD-20-0026 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Vong P, Ouled-Haddou H, Garçon L: Histone Deacetylases Function in the Control of Early Hematopoiesis and Erythropoiesis. Int J Mol Sci. 2022; 23(17): 9790. 10.3390/ijms23179790 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. AbdElmoniem N, Abdallah MH, Mukhtar RM, et al. : Identification of Novel Natural Dual HDAC and Hsp90 Inhibitors for Metastatic TNBC Using e-Pharmacophore Modeling, Molecular Docking, and Molecular Dynamics Studies. Molecules. 2023; 28(4): 1771. 10.3390/molecules28041771 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Wang Y, Fiskus W, Chong DG, et al. : Cotreatment with panobinostat and JAK2 inhibitor TG101209 attenuates JAK2V617F levels and signaling and exerts synergistic cytotoxic effects against human myeloproliferative neoplastic cells. Blood. 2009; 114(24): 5024–33. 10.1182/blood-2009-05-222133 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Harrison C, Heidel FH, Vannucchi AM, et al. : A Phase Ib Dose-finding Study of Panobinostat and Ruxolitinib in Myelofibrosis. HemaSphere. 2022; 6(8): e757. 10.1097/HS9.0000000000000757 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Yue L, Sharma V, Horvat NP, et al. : HDAC11 deficiency disrupts oncogene-induced hematopoiesis in myeloproliferative neoplasms. Blood. 2020; 135(3): 191–207. 10.1182/blood.2019895326 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Khan I, Huang Z, Wen Q, et al. : AKT is a therapeutic target in myeloproliferative neoplasms. Leukemia. 2013; 27(9): 1882–90. 10.1038/leu.2013.167 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Bogani C, Bartalucci N, Martinelli S, et al. : mTOR inhibitors alone and in combination with JAK2 inhibitors effectively inhibit cells of myeloproliferative neoplasms. PLoS One. 2013; 8(1): e54826. 10.1371/journal.pone.0054826 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Choong ML, Pecquet C, Pendharkar V, et al. : Combination treatment for myeloproliferative neoplasms using JAK and pan-class I PI3K inhibitors. J Cell Mol Med. 2013; 17(11): 1397–409. 10.1111/jcmm.12156 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Mascarenhas JO, Verstovsek S: The clinical dilemma of JAK inhibitor failure in myelofibrosis: Predictive characteristics and outcomes. Cancer. 2022; 128(14): 2717–27. 10.1002/cncr.34222 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Han X, Mei Y, Mishra RK, et al. : Targeting pleckstrin-2/Akt signaling reduces proliferation in myeloproliferative neoplasm models. J Clin Invest. 2023; 133(6): e159638. 10.1172/JCI159638 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Bianchi E, Rontauroli S, Tavernari L, et al. : Inhibition of ERK1/2 signaling prevents bone marrow fibrosis by reducing osteopontin plasma levels in a myelofibrosis mouse model. Leukemia. 2023; 37(5): 1068–1079. 10.1038/s41375-023-01867-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Rampal RK, Pinzon-Ortiz M, Somasundara AVH, et al. : Therapeutic Efficacy of Combined JAK1/2, Pan-PIM, and CDK4/6 Inhibition in Myeloproliferative Neoplasms. Clin Cancer Res. 2021; 27(12): 3456–68. 10.1158/1078-0432.CCR-20-4898 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Dutta A, Nath D, Yang Y, et al. : Genetic ablation of Pim1 or pharmacologic inhibition with TP-3654 ameliorates myelofibrosis in murine models. Leukemia. 2022; 36(3): 746–59. 10.1038/s41375-021-01464-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Mazzacurati L, Collins RJ, Pandey G, et al. : The pan-PIM inhibitor INCB053914 displays potent synergy in combination with ruxolitinib in models of MPN. Blood Adv. 2019; 3(22): 3503–14. 10.1182/bloodadvances.2019000260 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Marubayashi S, Koppikar P, Taldone T, et al. : HSP90 is a therapeutic target in JAK2-dependent myeloproliferative neoplasms in mice and humans. J Clin Invest. 2010; 120(10): 3578–93. 10.1172/JCI42442 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Bhagwat N, Koppikar P, Keller M, et al. : Improved targeting of JAK2 leads to increased therapeutic efficacy in myeloproliferative neoplasms. Blood. 2014; 123(13): 2075–83. 10.1182/blood-2014-01-547760 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Hobbs G, Litvin R, Ahn J, et al. : UY922, a Heat Shock Protein 90 (Hsp90) Inhibitor, Demonstrates Activity in Patients with Myeloproliferative Neoplasms (MPNs). Blood. 2015; 126(23): 4075. 10.1182/blood.V126.23.4075.4075 [DOI] [Google Scholar]
- 98. Uras IZ, Maurer B, Nivarthi H, et al. : CDK6 coordinates JAK2V617F mutant MPN via NF-κB and apoptotic networks. Blood. 2019; 133(15): 1677–90. 10.1182/blood-2018-08-872648 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Dutta A, Nath D, Yang Y, et al. : CDK6 Is a Therapeutic Target in Myelofibrosis. Cancer Res. 2021; 81(16): 4332–45. 10.1158/0008-5472.CAN-21-0590 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 100. De Dominici M, Porazzi P, Xiao Y, et al. : Selective inhibition of Ph-positive ALL cell growth through kinase-dependent and -independent effects by CDK6-specific PROTACs. Blood. 2020; 135(18): 1560–73. 10.1182/blood.2019003604 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Wen Q, Goldenson B, Silver SJ, et al. : Identification of regulators of polyploidization presents therapeutic targets for treatment of AMKL. Cell. 2012; 150(3): 575–89. 10.1016/j.cell.2012.06.032 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Wen QJ, Yang Q, Goldenson B, et al. : Targeting megakaryocytic-induced fibrosis in myeloproliferative neoplasms by AURKA inhibition. Nat Med. 2015; 21(12): 1473–80. 10.1038/nm.3995 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 103. Gangat N, Marinaccio C, Swords R, et al. : Aurora Kinase A Inhibition Provides Clinical Benefit, Normalizes Megakaryocytes, and Reduces Bone Marrow Fibrosis in Patients with Myelofibrosis: A Phase I Trial. Clin Cancer Res. 2019; 25(16): 4898–906. 10.1158/1078-0432.CCR-19-1005 [DOI] [PubMed] [Google Scholar]
- 104. Waibel M, Solomon VS, Knight DA, et al. : Combined targeting of JAK2 and Bcl-2/Bcl-xL to cure mutant JAK2-driven malignancies and overcome acquired resistance to JAK2 inhibitors. Cell Rep. 2013; 5(4): 1047–59. 10.1016/j.celrep.2013.10.038 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 105. Waksal JA, Mascarenhas J: Novel Therapies in Myelofibrosis: Beyond JAK Inhibitors. Curr Hematol Malig Rep. 2022; 17(5): 140–54. 10.1007/s11899-022-00671-7 [DOI] [PubMed] [Google Scholar]
- 106. Nakatake M, Monte-Mor B, Debili N, et al. : JAK2V617F negatively regulates p53 stabilization by enhancing MDM2 via La expression in myeloproliferative neoplasms. Oncogene. 2012; 31(10): 1323–33. 10.1038/onc.2011.313 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 107. Verstovsek S, Al-Ali HK, Mascarenhas J, et al. : BOREAS: a global, phase III study of the MDM2 inhibitor navtemadlin (KRT-232) in relapsed/refractory myelofibrosis. Future Oncol. 2022. 10.2217/fon-2022-0901 [DOI] [PubMed] [Google Scholar]
- 108. Marcellino BK, Clementelli C, Beaumont K, et al. : Dual Targeting of HDM2 and PPM1D As a Novel Therapeutic Approach for Myelofibrosis. Blood. 2022; 140(Supplement 1): 797–8. 10.1182/blood-2022-17023236006672 [DOI] [Google Scholar]
- 109. Maslah N, Verger E, Giraudier S, et al. : Single-cell analysis reveals selection of TP53-mutated clones after MDM2 inhibition. Blood Adv. 2022; 6(9): 2813–23. 10.1182/bloodadvances.2021005867 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Yan D, Pomicter AD, Tantravahi S, et al. : Nuclear-Cytoplasmic Transport Is a Therapeutic Target in Myelofibrosis. Clin Cancer Res. 2019; 25(7): 2323–35. 10.1158/1078-0432.CCR-18-0959 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Tantravahi SK, Kim SJ, Sundar D, et al. : A Phase 2 Study to Evaluate the Efficacy and Safety of Selinexor in Patients with Myelofibrosis Refractory or Intolerant to JAK Inhibitors. Blood. 2021; 138(Supplement 1): 143. 10.1182/blood-2021-153940 [DOI] [Google Scholar]
- 112. Craver BM, Nguyen TK, Nguyen J, et al. : The SMAC mimetic LCL-161 selectively targets JAK2V617F mutant cells. Exp Hematol Oncol. 2020; 9: 1. 10.1186/s40164-019-0157-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Pemmaraju N, Carter BZ, Bose P, et al. : Final results of a phase 2 clinical trial of LCL161, an oral SMAC mimetic for patients with myelofibrosis. Blood Adv. 2021; 5(16): 3163–73. 10.1182/bloodadvances.2020003829 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Mascarenhas J, Li T, Sandy L, et al. : Anti-transforming growth factor-β therapy in patients with myelofibrosis. Leuk Lymphoma. 2014; 55(2): 450–2. 10.3109/10428194.2013.805329 [DOI] [PubMed] [Google Scholar]
- 115. Varricchio L, Iancu-Rubin C, Upadhyaya B, et al. : TGF-β1 protein trap AVID200 beneficially affects hematopoiesis and bone marrow fibrosis in myelofibrosis. JCI Insight. 2021; 6(18): e145651. 10.1172/jci.insight.145651 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Rai S, Grockowiak E, Hansen N, et al. : Inhibition of interleukin-1β reduces myelofibrosis and osteosclerosis in mice with JAK2-V617F driven myeloproliferative neoplasm. Nat Commun. 2022; 13(1): 5346. 10.1038/s41467-022-32927-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Arranz L, Sánchez-Aguilera A, Martín-Pérez D, et al. : Neuropathy of haematopoietic stem cell niche is essential for myeloproliferative neoplasms. Nature. 2014; 512(7512): 78–81. 10.1038/nature13383 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 118. rexler B, Passweg JR, Tzankov A, et al. : The sympathomimetic agonist mirabegron did not lower JAK2-V617F allele burden, but restored nestin-positive cells and reduced reticulin fibrosis in patients with myeloproliferative neoplasms: results of phase II study SAKK 33/14. Haematologica. 2019; 104(4): 710–6. 10.3324/haematol.2018.200014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Pemmaraju N, Verstovsek S, Mesa R, Gupta V, Garcia JS, Scandura JM, et al. : Defining disease modification in myelofibrosis in the era of targeted therapy. Cancer. 2022; 128(13): 2420–32. 10.1002/cncr.34205 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Verstovsek S, Foltz L, Gupta V, et al. : Safety and efficacy of zinpentraxin alfa as monotherapy or in combination with ruxolitinib in myelofibrosis: stage I of a phase II trial. Haematologica. 2023. 10.3324/haematol.2022.282411 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Leiva O, Ng SK, Matsuura S, et al. : Novel lysyl oxidase inhibitors attenuate hallmarks of primary myelofibrosis in mice. Int J Hematol. 2019; 110(6): 699–708. 10.1007/s12185-019-02751-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Verstovsek S, Savona MR, Mesa RA, et al. : A phase 2 study of simtuzumab in patients with primary, post-polycythaemia vera or post-essential thrombocythaemia myelofibrosis. Br J Haematol. 2017; 176(6): 939–49. 10.1111/bjh.14501 [DOI] [PubMed] [Google Scholar]
- 123. Verstovsek S, Mascarenhas J, Rampal RK, et al. : Mylox-1: An Open-Label, Phase IIa Study of the Safety, Tolerability, Pharmacokinetics and Pharmacodynamics of Oral LOXL2 Inhibitor, GB2064, in Myelofibrosis. Blood. 2022; 140(Supplement 1): 3868–70. 10.1182/blood-2022-167496 [DOI] [Google Scholar]
- 124. Verachi P, Gobbo F, Martelli F, et al. : Preclinical studies on the use of a P-selectin-blocking monoclonal antibody to halt progression of myelofibrosis in the Gata1low mouse model. Exp Hematol. 2023; 117: 43–61. 10.1016/j.exphem.2022.09.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Passamonti F, Harrison CN, Mesa RA, et al. : Anemia in myelofibrosis: Current and emerging treatment options. Crit Rev Oncol Hematol. 2022; 180: 103862. 10.1016/j.critrevonc.2022.103862 [DOI] [PubMed] [Google Scholar]
- 126. Dussiot M, Maciel TT, Fricot A, et al. : An activin receptor IIA ligand trap corrects ineffective erythropoiesis in β-thalassemia. Nat Med. 2014; 20(4): 398–407. 10.1038/nm.3468 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 127. Hobbs G, Cimen Bozkus C, Moshier E, et al. : PD-1 inhibition in advanced myeloproliferative neoplasms. Blood Adv. 2021; 5(23): 5086–97. 10.1182/bloodadvances.2021005491 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Gigoux M, Holmström MO, Zappasodi R, et al. : Calreticulin mutant myeloproliferative neoplasms induce MHC-I skewing, which can be overcome by an optimized peptide cancer vaccine. Sci Transl Med. 2022; 14(649): eaba4380. 10.1126/scitranslmed.aba4380 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Liu P, Zhao L, Loos F, et al. : Immunosuppression by Mutated Calreticulin Released from Malignant Cells. Mol Cell. 2020; 77(4): 748–60.e9. 10.1016/j.molcel.2019.11.004 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 130. Fucikova J, Spisek R, Kroemer G, et al. : Calreticulin and cancer. Cell Res. 2021; 31(1): 5–16. 10.1038/s41422-020-0383-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Handlos Grauslund J, Holmström MO, Jørgensen NG, 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. 10.3389/fonc.2021.637420 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Psaila B, Wang G, Rodriguez-Meira A, et al. : Single-Cell Analyses Reveal Megakaryocyte-Biased Hematopoiesis in Myelofibrosis and Identify Mutant Clone-Specific Targets. Mol Cell. 2020; 78(3): 477–92.e8. 10.1016/j.molcel.2020.04.008 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 133. Yacoub A, Patnaik MM, Ali H, et al. : A Phase 1/2 Study of Single Agent Tagraxofusp, a First-in-Class CD123-Targeted Therapy, in Patients with Myelofibrosis That Is Relapsed/Refractory Following JAK Inhibitor Therapy. Blood. 2021; 138(Supplement1): 140. 10.1182/blood-2021-145276 [DOI] [Google Scholar]
- 134. Jutzi JS, Marneth AE, Ciboddo M, et al. : Whole-genome CRISPR screening identifies N-glycosylation as a genetic and therapeutic vulnerability in CALR-mutant MPNs. Blood. 2022; 140(11): 1291–304. 10.1182/blood.2022015629 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Rivera JF, Baral AJ, Nadat F, et al. : Zinc-dependent multimerization of mutant calreticulin is required for MPL binding and MPN pathogenesis. Blood Adv. 2021; 5(7): 1922–32. 10.1182/bloodadvances.2020002402 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Tvorogov D, Thompson-Peach CAL, Foßelteder J, et al. : Targeting human CALR-mutated MPN progenitors with a neoepitope-directed monoclonal antibody. EMBO Rep. 2022; 23(4): e52904. 10.15252/embr.202152904 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 137. Reis E, Buonpane R, Celik, H, et al. : Discovery of INCA033989, a Monoclonal Antibody That Selectively Antagonizes Mutant Calreticulin Oncogenic Function in Myeloproliferative Neoplasms (MPNs). Blood. 2022; 140(Supplement 1): 14–5. 10.1182/blood-2022-159435 [DOI] [Google Scholar]
- 138. Kihara Y, Araki M, Imai M, et al. : Therapeutic Potential of an Antibody Targeting the Cleaved Form of Mutant Calreticulin in Myeloproliferative Neoplasms. Blood. 2020; 136(Suppl 1): 9–10. 10.1182/blood-2020-141159 [DOI] [Google Scholar]
- 139. Achyutuni S, Nivarthi H, Majoros A, et al. : Hematopoietic expression of a chimeric murine-human CALR oncoprotein allows the assessment of anti-CALR antibody immunotherapies in vivo. Am J Hematol. 2021; 96(6): 698–707. 10.1002/ajh.26171 [DOI] [PubMed] [Google Scholar]
- 140. Pronier E, Cifani P, Merlinsky TR, et al. : Targeting the CALR interactome in myeloproliferative neoplasms. JCI insight. 2018; 3(22): e122703. 10.1172/jci.insight.122703 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Meyer SC, Keller MD, Chiu S, et al. : CHZ868, a Type II JAK2 Inhibitor, Reverses Type I JAK Inhibitor Persistence and Demonstrates Efficacy in Myeloproliferative Neoplasms. Cancer cell. 2015; 28(1): 15–28. 10.1016/j.ccell.2015.06.006 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 142. Rai S, Stetka J, Usart M, et al. : The Second Generation Type II JAK2 Inhibitor, AJ1-10502, Demonstrates Enhanced Selectivity, Improved Therapeutic Efficacy and Reduced Mutant Cell Fraction Compared to Type I JAK2 Inhibitors in Models of Myeloproliferative Neoplasms (MPNs). Blood. 2022; 140(Supplement 1): 6722–23. 10.1182/blood-2022-162621 [DOI] [Google Scholar]
- 143. Leroy E, Constantinescu SN: Rethinking JAK2 inhibition: towards novel strategies of more specific and versatile Janus kinase inhibition. Leukemia. 2017; 31(5): 1023–38. 10.1038/leu.2017.43 [DOI] [PubMed] [Google Scholar]
- 144. Leroy E, Balligand T, Pecquet C, et al. : Differential effect of inhibitory strategies of the V617 mutant of JAK2 on cytokine receptor signaling. J Allergy Clin Immunol. 2019; 144(1): 224–35. 10.1016/j.jaci.2018.12.1023 [DOI] [PubMed] [Google Scholar]
- 145. Wilmes S, Hafer M, Vuorio J, et al. : Mechanism of homodimeric cytokine receptor activation and dysregulation by oncogenic mutations. Science. 2020; 367(6478): 643–52. 10.1126/science.aaw3242 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 146. Glassman CR, Tsutsumi N, Saxton RA, et al. : Structure of a Janus kinase cytokine receptor complex reveals the basis for dimeric activation. Science. 2022; 376(6589): 163–9. 10.1126/science.abn8933 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 147. Chang Y, Min J, Jarusiewicz JA, et al. : Degradation of Janus kinases in CRLF2-rearranged acute lymphoblastic leukemia. Blood. 2021; 138(23): 2313–26. 10.1182/blood.2020006846 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Moraga I, Wernig G, Wilmes S, et al. : Tuning cytokine receptor signaling by re-orienting dimer geometry with surrogate ligands. Cell. 2015; 160(6): 1196–208. 10.1016/j.cell.2015.02.011 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation