USE OF AN ERK INHIBITOR FOR THE TREATMENT OF MYELOFIBROSIS

Information

  • Patent Application
  • 20240000777
  • Publication Number
    20240000777
  • Date Filed
    October 07, 2021
    2 years ago
  • Date Published
    January 04, 2024
    4 months ago
Abstract
The invention relates to the use of an ERK inhibitor in the treatment of myelofibrosis (MF). The invention also relates to a pharmaceutical combination comprising a) an ERK inhibitor and b) at least one further therapeutic agent, preferably ruxolitinib or a pharmaceutically acceptable salt thereof.
Description

The present invention provides uses of ERK inhibitors and combinations thereof, and their uses in the treatment of a disease or disorder as described herein, or methods of treating a disease or disorder as described herein.


FIELD OF THE INVENTION

The present invention provides the use of an ERK inhibitor, e.g. 4-(3-amino-6-((1S,3S,4S)-3-fluoro-4-hydroxycyclohexyl)pyrazin-2-yl)-N—((S)-1-(3-bromo-5-fluorophenyl)-2-(methylamino)ethyl)-2-fluorobenzamide (Compound A), in the treatment of myeloproliferative neoplasms (MPNs), including the treatment of myelofibrosis (MF), essential thrombocythemia (ET) and/or polycythemia vera (PV). It also provides a pharmaceutical combination comprising an ERK 1/2 inhibitor, e.g. Compound A, and a JAK inhibitor, in particular a JAK1/2 inhibitor such as ruxolitinib, and to the use of this combination in the treatment of myeloproliferative neoplasms (MPNs), including the treatment of myelofibrosis (MF), essential thrombocythemia (ET) and/or polycythemia vera (PV).


The invention also provides a pharmaceutical combination or a pharmaceutical composition for the treatment of MF comprising a) a ERK inhibitor and b) at least one further therapeutic agent.


The invention also provides methods of treating a disease or disorder, in particular myeloproliferative neoplasms (MPNs), including the treatment of myelofibrosis (MF), essential thrombocythemia (ET) and/or polycythemia vera (PV), in a patient in need thereof comprising administering to said patient a jointly therapeutically effective amount of pharmaceutical combinations or pharmaceutical compositions as provided herein. Also provided are uses of such combinations or compositions for the treatment of a disease or disorder, in particular myeloproliferative neoplasms (MPNs), including the treatment of myelofibrosis (MF), essential thrombocythemia (ET) and/or polycythemia vera (PV). The present invention also provides pharmaceutical compositions comprising such combinations and commercial packages thereto, and their uses in treating a disease or a disorder as described herein.


BACKGROUND

Myeloproliferative neoplasms (MPNs) are a unique and heterogeneous group of homeopathies characterized by proliferation and accumulation of mature myeloid cells. MPNs, include myelofibrosis (MF), essential thrombocythemia (ET) and polycythemia vera (PV). Importantly, MF is the most severe form of Philadelphia chromosome-negative (i.e. BCR-ABL1-negative) myeloproliferative neoplasms, with a prevalence estimated to be 2.2 per 100,000 population. MF can present as a de novo disorder as a primary hematologic malignancy, primary myelofibrosis (PMF) or evolve from previous myeloproliferative neoplasms, namely: PV, post-PV MF (PPV-MF), ET, or post-ET MF (PET-MF). The range of reported frequencies for post-PV MF are 4.9-6% at 10 years and 6-14% at 15 years, respectively, and 0.8-4.9% for post-ET MF at 10 years and 4-11% at 15 years, respectively (S Cerquozzi and A Tefferi, Blood Cancer Journal (2015) 5, e366).


Regardless of whether MF developed from PV, ET or as a primary disorder, it is characterized by a clonal stem cell proliferation associated with production of elevated levels of several inflammatory and proangiogenic cytokines resulting in a bone marrow stromal reaction that includes varying degrees of reticuline and/or collagen fibrosis, osteoschlerosis and angiogenesis, some degree of megakaryocyte atypia and a peripheral blood smear showing a leucoerythroblastic pattern with varying degrees of circulating progenitor cells. The abnormal bone marrow milieu results in release of hematopoietic stem cells into the blood, extramedullary hematopoiesis, and organomegaly at these sites. Clinically, MF is characterized by progressive anemia, leukopenia or leukocytosis, thrombocytopenia or thrombocythemia and multi-organ extramedullary hematopoiesis, which most prominently involves the spleen leading to massive splenomegaly, severe constitutional symptoms, a hypermetabolic state, cachexia, and premature death.


A considerable number of cytokine and growth factor receptors utilize non-receptor tyrosine kinases, the Janus kinases (JAK), to transmit extracellular ligand binding into an intracellular response. For example, erythropoietin, thrombopoietin and granulocyte monocyte colony stimulating factor are all known to signal through receptors that utilize JAK2. JAK activate a number of downstream pathways implicated in proliferation and survival, including the STATs (signal transducers and activators of transcription), a family of important latent transcription factors.


Myelofibrosis is now known to be a clonal stem cell disease characterized by molecular (JAK2V617F, MPLW515L/K) and cytogenetic (13q-,20q-) markers (Pikman Y, Lee B H, Mercher T, et al. PLoS Med. 2006; 3(7):e270; Scott L M, Tong W, Levine R L, et al. N Engl J Med. 2007; 356:459-468). The JAK2V617F mutation has been identified in over 95% of patients with PV and approximately 50% of patients with ET and PMF. Furthermore, in a preclinical setting, animal studies have demonstrated that this mutation can lead to an MF-like syndrome. The JAK2V617F mutation alters the JAK2 tyrosine kinase making it constitutively active. As a result, polycythemia, thrombocythemia and leukocytosis can develop independently from growth factor regulation. Even in patients lacking a confirmed JAK2 mutation, the detection of STAT activation suggests dysregulated JAK activity. In fact, regardless of the mutational status of JAK2, the malignant cells appear to retain their responsiveness to JAK activating cytokines and/or growth factors; hence, they may benefit from JAK inhibition. Although several JAK inhibitors, including ruxolitinib (brand name Jakavi) have been approved for the treatment of MF, they have only demonstrated an effect in the treatment of symptoms. Progression of the disease is not halted and eventually patients may die prematurely.


Patients with MF have shortened survival (median survival is 6.5 years) and greatly compromised quality of life (QoL). Contributing factors for shortened survival include leukemic transformation and thrombohemorrhagic complications and for the compromised quality of life severe anemia (often requiring red blood cell (RBC) transfusions), symptomatic enlargement of the spleen and liver, substantial MF-associated symptoms burden (MF-SB), and cachexia (Tefferi and Barbui 2019).


The only potential curative treatment for MF is allogeneic hematopoietic stem cell transplantation (ASCT), for which the great majority of patients are ineligible. Therefore, treatment options remain primarily palliative and aimed at controlling disease symptoms, complications and improving the patient's QoL. The therapeutic landscape of MF has changed with the discovery of the V617F mutation of the Janus kinase JAK2 gene present in 60% of patients with PMF or PET-MF and in 95% of patients with PPV-MF, triggering the development of molecular targeted therapy for MF (Cervantes 2014). JAK play an important role in signal transduction following cytokine and growth factor binding to their receptors. Aberrant activation of JAK has been associated with increased malignant cell proliferation and survival (Valentino and Pierre 2006). JAK activate a number of downstream signaling pathways implicated in the proliferation and survival of malignant cells including members of the Signal Transducer and Activator of Transcriptions (STAT) family of transcription factors.


JAK inhibitors were developed to target JAK2 thereby inhibiting JAK signaling. Ruxolitinib, as all agents of this class, mainly inhibits dysregulated JAK-STAT signaling present in all MF patients irrespective of their JAK2 mutational status, but is not selective for the mutated JAK2, which explains its efficacy in both JAK2-positive and -negative MF. Ruxolitinib is highly effective in reducing the spleen size and controlling the symptoms of MF, with this resulting in a marked improvement in the patient's QoL (Cervantes et al 2016). Ruxolitinib is the only JAK inhibitor that has been granted a marketing authorization, as a single agent, for the treatment of patients with PMF, PPV-MF or PET-MF and for the treatment of patients with PV who are resistant to or intolerant to hydroxyurea. Ruxolitinib is the only approved pharmacological treatment for MF patients with splenomegaly and/or clinical symptoms and is considered the standard of care (SoC). Although ruxolitinib has changed the treatment paradigm of MF patients, there is no clear indication of its disease-modifying effect (Cervantes 2014) and therapy-related anemia is often an anticipated downside (Naymagon and Mascarenhas 2017, Mead et al 2015). While ruxolitinib demonstrates improvements in splenomegaly and constitutional symptoms, it has not been shown to improve anemia.


Current treatment options post-JAK inhibitors are limited in their efficacy, durability and tolerability. Multiple efforts are currently ongoing to improve the outcome of patients with MF post JAK inhibitors identifying new agents or combinations, such as those targeting cellular metabolic and apoptotic pathways, cell cycle and immune therapy. There remains a high unmet medical need to finding new and efficacious therapeutic options for advancing the treatment of myelofibrosis. There is also a need for targeted therapy that is safe and/or well tolerated. For example, there is a need for a therapy that would help to overcome the side effects such as anemia, which are associated with standard of care such as monotherapy with ruxolitinib.


WO/2015/066188 discloses inhibitors of ERK1/2 such as compound A, also known as rineterkib, as being useful in treating diseases such as cancer that are associated with excessive activity of ERK1 and/or ERK2. However, it does not specifically disclose the use of ERK1/2 inhibitors in the treatment of myelofibrosis or in combination with a JAK inhibitor such as ruxolitinib.


SUMMARY OF THE INVENTION

Inhibition of JAK2 and ERK1/2 with a combination of ruxolitinib and several ERK inhibitors (such as Compound A and MK-8535) inhibited proliferation of Jak2V617F Ba/F3 cells. Treatment of mice competitively transplanted with Jak2V617F and wild-type BM with the combinations of the invention corrected erythrocytosis and splenomegaly. Longer-term treatment was able to induce clone reductions. BM fibrosis was profoundly decreased in MPLW515L-driven MPN to an extent not seen with JAK2 inhibitor monotherapy. Myeloid colony formation from JAK2V617F patients' CD34+ blood and BM was dose-dependently inhibited by combined JAK2/ERK1/2 inhibition in PV, ET and MF subsets. The fitness of the MPN clone was found to be decreased.


It was found that an ERK 1/2 inhibitor such as Compound A as defined below, and especially when used in combination with a JAK inhibitor, and in particular a JAK1/2 inhibitor such as ruxolitinib, significantly normalizes splenomegaly, polycythemias and Hematocrit in a MF mouse model. The combination of the ERK 1/2 inhibitor and the JAK 1/2 inhibitor was also found to be well tolerated in a MF mouse model.


The present invention therefore provides a novel therapy, which may deliver clinical benefit to a patient suffering from MPNs such as MF and/or PV. In particular, the present invention may provide an improvement of anemia and progression free survival for such patients.


The present invention thus provides a medicament for the treatment of myelofibrosis. The present invention is based on the inventors' surprising finding that an ERK1/2 inhibitor is useful in the treatment of myelofibrosis in a patient.


The present invention thus provides combinations, methods, or compounds for use in the treatment of a disorder or disease or for use in the alleviation of a symptom or symptoms associated with the disorder or disease as described herein.


The present invention is also based on finding that an ERK1/2 inhibitor in combination with at least one further therapeutic agent such as ruxolitinib is useful in the treatment of myelofibrosis in a patient.


In an embodiment, the ERK1/2 inhibitor is selected from Compound A (rineterkib), BVD-523 (ulixertinib), GDC-0994, KO-947, Vtx-11e, SCH-772984, MK2853, LY3214996, BVD-523, SCH-722984, LY3214996, SCH-900353, AEZS-140, AEZS-131, AEZS-136, RG-7842 CC-90003, KIN-4050, and combinations thereof.


In an embodiment, the ERK1/2 inhibitor is Compound A (rineterkib), BVD-523 (ulixertinib), SCH-772984, MK2853, SCH-722984, or DEL22379.


In an embodiment, the ERK1/2 inhibitor is Compound A (rineterkib), SCH-772984, MK2853, or SCH-722984.


In an embodiment, the ERK1/2 inhibitor is a compound having the structure of Formula (I)




embedded image


4-(3-amino-6-((1S,3S,4S)-3-fluoro-4-hydroxycyclohexyl)pyrazin-2-yl)-N—((S)-1-(3-bromo-5-fluorophenyl)-2-(methylamino)ethyl)-2-fluorobenzamide (“Compound A”) or a pharmaceutically acceptable salt thereof, for example the hydrochloride salt thereof.


In an embodiment, the JAK inhibitor is a JAK1/2 inhibitor.


In an embodiment, the JAK inhibitor is ruxolitinib or pharmaceutically acceptable salt thereof, for example the phosphate salt thereof.


In an embodiment, the JAK inhibitor is itacitinib, or pharmaceutically acceptable salt thereof.


In an embodiment, the JAK inhibitor is momelotinib, or pharmaceutically acceptable salt thereof.


In an embodiment, Compound A and the JAK inhibitor are in the same pharmaceutical formulation.


In another embodiment, Compound A and the JAK inhibitor are in separate pharmaceutical formulations.


In a further embodiment, the pharmaceutical combination is for use in simultaneous or sequential administration.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1A depicts the antiproliferative effect of Compound A (LTT462), ruxolitinib (Rux), and ruxolitinib (Rux) and Compound A (LTT462) in the Ba/F3 EpoR JAK2V617F cell line.



FIG. 1B depicts the antiproliferative effect of Compound A (LTT462), ruxolitinib (Rux), and ruxolitinib (Rux) and Compound A (LTT462) in the SET2 cell line.



FIG. 2A depicts the effects of vehicle, ruxolitinib (Rux), Compound A (LTT462), and ruxolitinib (Rux) and Compound A (LTT462) on ERK signaling in JAK2V617F PV/MF mouse model.



FIG. 2B depicts the effects of vehicle, ruxolitinib (Rux), Compound A (LTT462), and ruxolitinib (Rux) and Compound A (LTT462) on hematocrit levels in JAK2V617F PV/MF mouse model.



FIG. 2C depicts the effects of vehicle, ruxolitinib (Rux), Compound A (LTT462), and ruxolitinib (Rux) and Compound A (LTT462) on spleen weight (as a percentage of body weight) in JAK2V617F PV/MF mouse model.



FIG. 2D depicts the effects of vehicle, ruxolitinib (Rux), Compound A (LTT462), and ruxolitinib (Rux) and Compound A (LTT462) on white blood count (WBC count in in MPLW515L mice.



FIG. 3A depicts the activity of vehicle, ruxolitinib (Rux), Compound A (LTT462), and ruxolitinib (Rux) and Compound A (LTT462) on the reduction of elevated hematocrit in JAK2V617F PV/MF mouse model.



FIG. 3B depicts the activity of vehicle, ruxolitinib (Rux), Compound A (LTT462), and ruxolitinib (Rux) and Compound A (LTT462) on normalizing splenomegaly in JAK2V617F PV/MF mouse model.



FIG. 4A depicts the tolerability of the treatments with vehicle, ruxolitinib (Rux), Compound A (LTT462), and ruxolitinib (Rux) and Compound A (LTT462), as indicated by body weight change over time.



FIG. 4B depicts the tolerability of the treatments with vehicle, ruxolitinib (Rux), Compound A (LTT462), and ruxolitinib (Rux) and Compound A (LTT462), as indicated by bone marrow cellularity, indicating absence of bone marrow toxicity.



FIG. 4C depicts the tolerability of the treatments with vehicle, ruxolitinib (Rux), Compound A (LTT462), and ruxolitinib (Rux) and Compound A (LTT462), as indicated by normal WBC count.



FIG. 4D depicts the tolerability of the treatments with vehicle, ruxolitinib (Rux), Compound A (LTT462), and ruxolitinib (Rux) and Compound A (LTT462), as indicated by normal platelet count, indicating absence of thrombocytopenia.



FIG. 5A depicts in vitro activity of vehicle (Veh), Compound A (LTT462), ruxolitinib (Rux), and ruxolitinib (Rux) and Compound A (LTT462) in CD34+ peripheral blood mononuclear cells (PBMCs).



FIG. 5B depicts the activity of vehicle (Veh), Compound A (LTT462), ruxolitinib (Rux), and ruxolitinib (Rux) and Compound A (LTT462) on hematocrit levels in JAK2V617F mice.



FIG. 6 depicts the effects of Compound A (LTT462), ruxolitinib (Rux), and ruxolitinib (Rux) and Compound A (LTT462) on IC50 activity in EpoR Jak2V617F mutant and Jak2 wild type Ba/F3 cells.



FIG. 7A depicts the effects of ruxolitinib (ruxo) and ERK1/2 deletion in reducing mutant clone in Jak2V617F MPN mouse model.



FIG. 7B depicts the effects of ruxolitinib (ruxo) and ERK1/2 deletion in normalizing red blood cell count (RBC) in Jak2V617F MPN mouse model.





DETAILED DESCRIPTION OF THE INVENTION

Certain terms used herein are described below. Compounds or biological agents of the present invention are described using standard nomenclature. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which this invention belongs.


The term “combination,” “therapeutic combination,” or “pharmaceutical combination” as used herein refer to either a fixed combination in one dosage unit form, or non-fixed combination, or a kit of parts for the combined administration where two or more therapeutic agents may be administered independently, at the same time or separately within time intervals, especially where these time intervals allow that the combination partners show a cooperative, e.g., synergistic, effect.


The term “combination therapy” refers to the administration of two or more therapeutic agents to treat a therapeutic condition or disorder described in the present disclosure. Such administration encompasses co-administration of these therapeutic agents in a substantially simultaneous manner, such as in a single formulation having a fixed ratio of active ingredients or in separate formulations (e.g., capsules and/or intravenous formulations) for each active ingredient. In addition, such administration also encompasses use of each type of therapeutic agent in a sequential or separate manner, either at approximately the same time or at different times. Regardless of whether the active ingredients are administered as a single formulation or in separate formulations, the drugs are administered to the same patient as part of the same course of therapy. In any case, the treatment regimen will provide beneficial effects in treating the conditions or disorders described herein.


The term “JAK inhibitor” as used herein refers to a compound that selectively targets, decreases, or inhibits at least one activity of JAK.


The term “JAK1/2 inhibitor” as used herein refers to a compound that selectively targets, decreases, or inhibits the JAK 1 and JAK 2 tyrosine kinases.


The term “ERK inhibitor” as used herein refers to a compound that inhibits extracellular signal-regulated kinase (ERK).


The term “ERK 1/2 inhibitor” as used herein refers to a compound that inhibits ERK1 and/or ERK2 kinases.


The term “pharmaceutical composition” is defined herein to refer to a mixture or solution containing at least one therapeutic agent to be administered to a patient, e.g., a mammal or human, in order to prevent or treat a particular disease or condition affecting the mammal.


The term “pharmaceutically acceptable” as used herein refers to those compounds, biological agents (e.g., antibodies), materials, compositions and/or dosage forms, which are, within the scope of sound medical judgment, suitable for contact with the tissues of a warm-blooded animal, e.g., a mammal or human, without excessive toxicity, irritation, allergic response, and other problem complications commensurate with a reasonable benefit/risk ratio.


The terms “fixed combination,” “fixed dose,” and “single formulation” as used herein refers to a single carrier or vehicle or dosage form formulated to deliver an amount, which is jointly therapeutically effective for the treatment or prevention of cancer, of both therapeutic agents to a patient. The single vehicle is designed to deliver an amount of each of the agents, along with any pharmaceutically acceptable carriers or excipients. In some embodiments, the vehicle is a tablet, capsule, pill, or a patch. In other embodiments, the vehicle is a solution or a suspension.


The term “non-fixed combination,” “kit of parts,” and “separate formulations” means that at least one of the active ingredients is administered to a patient as a separate entity either simultaneously, concurrently, or sequentially with no specific time limits, wherein such administration provides therapeutically effective levels of the two active ingredients agents in the body of the patient in need thereof. The latter also applies to cocktail therapy, e.g., the administration of three or more active ingredients.


The term “unit dose” is used herein to mean simultaneous administration of both agents together, in one dosage form, to the patient being treated. In some embodiments, the unit dose is a single formulation. In certain embodiments, the unit dose includes one or more vehicles such that each vehicle includes an effective amount of at least one of the agents along with pharmaceutically acceptable carriers and excipients. In some embodiments, the unit dose is one or more tablets, capsules, pills, injections, infusions, patches, or the like, administered to the patient at the same time.


An “oral dosage form” includes a unit dosage form prescribed or intended for oral administration.


The term “treating” or “treatment” as used herein comprises a treatment relieving, reducing, or alleviating at least one symptom in a patient or effecting a delay of progression of a disease. For example, treatment can be the diminishment of one or several symptoms of a disorder or complete eradication of a disorder, such as cancer. Within the meaning of the present disclosure, the term “treat” also denotes to arrest, delay the onset (i.e., the period prior to clinical manifestation of a disease), and/or reduce the risk of developing or worsening a disease. The term “protect” is used herein to mean prevent, delay, or treat, or all, as appropriate, development, continuance or aggravation of a disease in a patient, e.g., a mammal or human. The term “prevent”, “preventing” or “prevention” as used herein comprises the prevention of at least one symptom associated with or caused by the state, disease or disorder being prevented.


The term “treatment” as used herein includes treatment of splenomegaly, treatment of hepatomegaly, treatment of thrombocytopenia, treatment of neutropenia, treatment of anemia, treatment of bone marrow fibrosis associated with MF, and treatment of a symptom associated with MPNs or a constitutional symptom associated with myelofibrosis.


The term “pharmaceutically effective amount,” “therapeutically effective amount,” or “clinically effective amount” of a combination of therapeutic agents is an amount sufficient to provide an observable or clinically significant improvement over the baseline clinically observable signs and symptoms of the disorders treated with the combination.


The term “jointly therapeutically active” or “joint therapeutic effect” as used herein means that the therapeutic agents can be given separately (in a chronologically staggered manner, especially a sequence-specific manner) in such time intervals that they prefer, in the warm-blooded animal, especially human, to be treated, still show an (preferably synergistic) interaction (joint therapeutic effect). Whether this is the case can, inter alia, be determined by following the blood levels of the compounds, showing that both compounds are present in the blood of the human to be treated at least during certain time intervals.


The terms “comprising” and “including” are used herein in their open-ended and non-limiting sense unless otherwise noted.


The terms “a” and “an” and “the” and similar references in the context of describing the invention (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. Where the plural form is used for compounds, biological agents, salts, and the like, this is taken to mean also a single compound, salt, or the like.


The terms “about” or “approximately” are generally understood by persons knowledgeable in the relevant subject area, but in certain circumstances can mean within 20%, within 10%, or within 5% of a given value or range. Alternatively, especially in biological systems, the term “about” means within about a log (i.e., an order of magnitude) or within a factor of two of a given value.


In particular, where a dosage is mentioned as ‘about’ a particular value, or a particular value (i.e. without the term “about” preceding that particular value, it is intended to include a range around the specified value of plus or minus 10%, or plus or minus 5%. As is customary in the art, dosages refer to the amount of the therapeutic agent in its free form. For example, when a dosage of 100 mg of Compound A is referred to, and Compound A is used as its hydrochloride salt, the amount of the therapeutic agent used is equivalent to 100 mg of the free form of Compound A.


In an embodiment, the ERK1/2 inhibitor is selected from Compound A (rineterkib), BVD-523 (ulixertinib), GDC-0994, KO-947, Vtx-11e, SCH-772984, MK2853, LY3214996, BVD-523, SCH-722984, LY3214996, SCH-900353, AEZS-140, AEZS-131, AEZS-136, RG-7842 CC-90003, KIN-4050, and combinations thereof.


In an embodiment, the ERK1/2 inhibitor is Compound A (rineterkib), BVD-523 (ulixertinib), SCH-772984, MK2853, SCH-722984, or DEL22379.


In an embodiment, the ERK1/2 inhibitor is Compound A (rineterkib), SCH-772984, MK2853, or SCH-722984.


In one preferred embodiment, the ERK1/2 inhibitor is Compound A, which is 4-(3-amino-6-((1S,3S,4S)-3-fluoro-4-hydroxycyclohexyl)pyrazin-2-yl)-N—((S)-1-(3-bromo-5-fluorophenyl)-2-(methylamino)ethyl)-2-fluorobenzamide:




embedded image


This compound is an inhibitor of ERK 1 and ERK 2. The compound is disclosed and its preparation described in published PCT patent application WO2015/066188 as example 184, which is incorporated herein by reference. This compound is also known as rineterkib. In some embodiments, this compound is used as its hydrochloride salt.


Any reference to “Compound A” herein is meant to include a reference to Compound A, or a pharmaceutically acceptable salt thereof, for example the hydrochloride salt thereof, unless context clearly indicates otherwise.


In one embodiment, an ERK inhibitor that can be used in the methods and combinations of the invention is BVD-523, also known as ulixertinib, which is (S)-4-(5-chloro-2-(isopropylamino)pyridin-4-yl)-N-(1-(3-chlorophenyl)-2-hydroxyethyl)-1H-pyrrole-2-carboxamide:




embedded image


Exemplary JAK inhibitors include, but are not limited to, ruxolitinib (Jakafi®); tofacitinib (CP690550); axitinib (AG013736, CAS 319460-85-0); 5-Chloro-N2-[(1S)-1-(5-fluoro-2-pyrimidinyl)ethyl]-N4-(5-methyl-1H-pyrazol-3-y)-l2,4-pyrimidinediamine (AZD1480, CAS 935666-88-9); (9E)-15-[2-(1-Pyrrolidinyl)ethoxy]-7,12,26-trioxa-19,21,24-triazatetracyclo[18.3.1.12,5.114,18]-hexacosa-1(24),2,4,9,14,16,18(25),20,22-nonaene (SB-1578, CAS 937273-04-6); momelotinib (CYT 387); baricitinib (INCB-028050 or LY-3009104); pacritinib (SB1518); (16E)-14-Methyl-20-oxa-5,7,14,27-tetraazatetracyclo[19.3.1.12,6.18,12]heptacosa-1(25),2,4,6(27),8,10,12(26),16,21,23-decaene (SB 1317); gandotinib (LY 2784544); and N,N-dicyclopropyl-4-[(1,5-dimethyl-1H-pyrazol-3-yl)amino]-6-ethyl-1,6-dihydro-1-methyl-imidazo[4,5-d]pyrrolo[2,3-b]pyridine-7-carboxamide (BMS 911543).


As used herein, “ruxolitinib” is the JAK1/JAK2 inhibitor (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile, also named 3(R)-Cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile, of formula:




embedded image


which can be prepared, for example, as described in WO2007/070514, which is incorporated herein by reference. As used herein, “ruxolitinib” refers to the free form, and any reference to “a pharmaceutically acceptable salt thereof” refers to “a pharmaceutically acceptable acid addition salt thereof”, in particular ruxolitinib phosphate, which can be prepared, for example, as described in WO2008/157208, which is incorporated herein by reference. Ruxolitinib is approved for the treatment of intermediate to high-risk myelofibrosis under the tradename Jakafi®/Jakavi®.


Ruxolitinib, or pharmaceutically acceptable salt thereof, in particular ruxolitinib phosphate, can be in a unit dosage form (e.g. tablet), which is administered orally.


As used herein, “ruxolitinib” is also intended to represent ruxolitinib, or a pharmaceutically acceptable salt thereof (for example the phosphate salt thereof), unless context clearly indicates otherwise.


In one embodiment, “ruxolitinib” is also intended to represent isotopically labeled forms. Isotopically labeled compounds have structures depicted by the formula above except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Isotopes that can be incorporated into ruxolitinib, for example, isotopes of hydrogen, namely the compound of formula:




embedded image


wherein each R1, R2, R3, R4, R5, R6, R7, R8, R9, R10, R11, R12, R13, R14, R15, R16 and R17 is independently selected from H or deuterium; provided that there is at least one deuterium present in the compound. In other embodiments, there are multiple deuterium atoms present in the compound. Suitable compounds are disclosed in U.S. Pat. No. 9,249,149 B2, which is hereby incorporated in its entirety.


In one preferred embodiment, a deuterated ruxolitinib is selected from the group consisting of




embedded image


or a pharmaceutically acceptable salt of any of the foregoing.


In a preferred embodiment, a deuterated ruxolitinib is




embedded image


or a pharmaceutically acceptable salt thereof.


As used herein, “itacitinib” refers to the JAK1/JAK2 inhibitor 2-(3-(4-(7H-pyrrolo(2,3-d)pyrimidin-4-yl)-1H-pyrazol-1-yl)-1-(1-(3-fluoro-2-(trifluoromethyl)isonicotinoyl)piperidin-4-yl)azetidin-3-yl)acetonitrile, also named 2-[1-[1-[3-fluoro-2-(trifluoromethyl)pyridine-4-carbonyl]piperidin-4-yl]-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)pyrazol-1-yl]azetidin-3-yl]acetonitrile of formula




embedded image


which can be prepared, for example, as described in WO2011/112662, which is incorporated herein by reference. As used herein, “itacitinib” refers to the free form, and any reference to “a pharmaceutically acceptable salt thereof” refers to “a pharmaceutically acceptable acid addition salt thereof”, in particular itacitinib adipate.


Treatment of Myeloproliferative Neoplasms (MPNs) and of Myelofibrosis

Myeloproliferative neoplasms (MPNs) are hematopoietic stem cell disorders characterized by excessive output of mature myeloid blood cells and an inherent risk for transformation to acute myeloid leukemia. MPN subtypes include; polycythemia vera (PV) primarily with polyglobulia, essential thrombocythemia (ET) with thrombocytosis, and myelofibrosis (MF) with an initial cell-rich phase followed by progressive bone marrow (BM) fibrosis and cytopenias. All of these MPN subsets share a common feature of dysregulated JAK2 signaling3, which is constitutively activated by somatic mutations in JAK2, the thrombopoietin receptor MPL or its stabilizing chaperone calreticulin (CALR)4.


Reference to the treatment of myeloproliferative neoplasms (MPNs) throughout this specification is therefore intended to include a disease or disorder selected from myelofibrosis (MF), essential thrombocythemia (ET), polycythemia vera (PV), and combinations thereof. Fpr example, the term “treatment” includes treatment of polycythemia vera (PV) primarily with polyglobulia, essential thrombocythemia (ET) with thrombocytosis, and myelofibrosis (MF) with an initial cell-rich phase followed by progressive bone marrow (BM) fibrosis and cytopenias.


In one aspect, the present invention provides an ERK1/2 inhibitor (e.g., Compound A) or a pharmaceutical acceptable salt thereof, alone or in combination with a JAK inhibitor, (e.g., ruxolitinib) or a pharmaceutical acceptable salt thereof, for use in the treatment of myeloproliferative neoplasms (MPNs).


In one aspect, the present invention provides an ERK1/2 inhibitor (e.g., Compound A) or a pharmaceutical acceptable salt thereof, alone or in combination with a JAK inhibitor, (e.g., ruxolitinib) or a pharmaceutical acceptable salt thereof, for use in the treatment of (i) myelofibrosis (MF), (ii) essential thrombocythemia (ET) or (iii) polycythemia vera (PV).


Myelofibrosis comprises primary myelofibrosis (PMF), post-essential thrombocythemia myelofibrosis (PET-MF) and post-polycythemia vera myelofibrosis (PPV-MF). Reference herein to the term “myelofibrosis” includes any one of a disorder selected from primary myelofibrosis (PMF), post-essential thrombocythemia myelofibrosis (PET-MF) and post-polycythemia vera myelofibrosis (PPV-MF). Suitably, myelofibrosis is PMF.


In one aspect the present invention provides an ERK1/2 inhibitor (e.g., Compound A) or a pharmaceutical acceptable salt thereof, alone or in combination with a JAK inhibitor, (e.g., ruxolitinib) or a pharmaceutical acceptable salt thereof, for use in the treatment of a Philadelphia-chromosome negative myeloproliferative neoplasm.


In another aspect, the present invention provides an ERK1/2 inhibitor (e.g., Compound A) or a pharmaceutical acceptable salt thereof, alone or in combination with a JAK inhibitor, (e.g., ruxolitinib) or a pharmaceutical acceptable salt thereof, for use in the treatment of polycythemia vera (PV).


In one further aspect, the present invention provides an ERK1/2 inhibitor (e.g., Compound A) or a pharmaceutical acceptable salt thereof, for use in the treatment of myelofibrosis (MF) in a patient. Alternatively, in one aspect, the present invention provides an ERK1/2 inhibitor (e.g., Compound A) or a pharmaceutical acceptable salt thereof, for use in the manufacture of a medicament for the treatment of myelofibrosis (MF) in a patient. Alternatively, in one aspect the present invention provides a method of treating myelofibrosis (MF) in a patient comprising the step of administering therapeutically effective amount of an ERK1/2 inhibitor (e.g., Compound A) or a pharmaceutical acceptable salt thereof, to said patient.


The term “primary myelofibrosis” (PMF), as used herein, is defined with reference to “The 2016 revision to the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia”, as published in Blood, 2016, 127:2391-2405. Primary myelofibrosis encompasses profibrotic/early primary myelofibrosis (prePMF) and overt primary myelofibrosis (overt PMF). Diagnosis of prePMF requires meeting the following 3 major criteria, and at least 1 minor criterion according to the 2016 WHO classification for prePMF in table A:









TABLE A





Criteria for diagnosis of prePMF















Major criteria (prePMF)


1. Megakaryocytic proliferation and atypia, without reticulin fibrosis >grade 1, accompanied by


increased age-adjusted BM cellularity, granulocytic proliferation, and often decreased


erythropoiesis


2. Not meeting the WHO criteria for BCR-ABL1+CML, PV, ET, myelodysplastic syndromes, or


other myeloid neoplasms


3. Presence of JAK2, CALR, or MPL mutation or in the absence of these mutations, presence


of another clonal marker (e.g., ASXL1, EZH2, TET2, IDH1/IDH2, SRSF2, SF3B1) are of help


in determining the clonal nature of the disease or absence of minor reactive bone marrow


(BM) reticulin fibrosis (Minor (grade 1) reticulin fibrosis secondary to infection, autoimmune


disorder or other chronic inflammatory conditions, hairy cell leukemia or other lymphoid


neoplasm, metastatic malignancy, or toxic (chronic) myelopathies)


Minor criteria (prePMF)


Presence of at least 1 of the following, confirmed in 2 consecutive determinations:


a. Anemia not attributed to a comorbid condition


b. Leukocytosis ≥11*109/L


c. Palpable splenomegaly


d. LDH increased to above upper normal limit of institutional reference range









Diagnosis of overt PMF requires meeting the following 3 major criteria, and at least 1 minor criterion according to the 2016 WHO classification for overt PMF in table B:









TABLE B





Criteria for diagnosis of overt PMF















Major criteria (overt PMF)


1. Presence of megakaryocytic proliferation and atypia, accompanied by either reticulin and/or


collagen fibrosis grades 2 or 3


2. Not meeting WHO criteria for ET, PV, BCR-ABL1+CML, myelodysplastic syndromes, or other


myeloid neoplasms


3. Presence of JAK2, CALR, or MPL mutation or in the absence of these mutations, presence of


another clonal marker (e.g., ASXL1, EZH2, TET2, IDH1/IDH2, SRSF2, SF3B1) or absence of


reactive myelofibrosis (BM fibrosis secondary to infection, autoimmune disorder, or other chronic


inflammatory conditions, hairy cell leukemia or other lymphoid neoplasm, metastatic malignancy,


or toxic (chronic) myelopathies)


Minor criteria (overt PMF)


Presence of at least 1 of the following, confirmed in 2 consecutive determinations:


a. Anemia not attributed to a comorbid condition


b. Leukocytosis ≥11*109/L


c. Palpable splenomegaly


d. LDH increased to above upper normal limit of institutional reference range


e. Leukoerythroblastosis









The term “bone marrow fibrosis”, as used herein, refers to bone marrow fibrosis graded according to the 2005 European consensus grading system (Thiele et. al., Haematologica, 2005, 90(8), 1128-1132, in particular as defined in Table 3 and FIG. 1 of page 1130 therein), such as:

    • “fibrosis grade 0”: scattered linear reticuline with no intersections (cross-overs) corresponding to normal bone marrow;
    • “fibrosis grade 1”: loose network of reticuline with many intersections, especially in perivascular areas;
    • “fibrosis grade 2”: diffuse and dense increase in reticuline with extensive intersections, occasionally with only focal bundles of collagen and/or focal osteoschlerosis;
    • “fibrosis grade 3”: diffuse and dense increase in reticuline with extensive intersections with coarse bundles of collagen, often associated with significant osteoschlerosis;
    • wherein the grading (i.e. grading of fiber density and quality) is made on the basis of bone marrow biopsy specimen assessment.


The term “essential thrombocythemia” (ET), as used herein, is defined with reference to “The 2016 revision to the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia”, as published in Blood, 2016, 127:2391-2405. The term “post-essential thrombocythemia myelofibrosis” (PET-MF), as used herein, refers to MF secondary to ET (i.e. MF arising as a progression of ET), wherein ET is as defined herein above. According to the IWG-MRT criteria (Barosi G et al, Leukemia (2008) 22, 437-438), criteria for diagnosing post-essential thrombocythemia myelofibrosis are:









TABLE C





Criteria for diagnosis of post-essential thrombocythemia myelofibrosis















Required criteria:


1. Documentation of a previous diagnosis of essential thrombocythemia as defined by the


WHO criteria


2. Bone marrow fibrosis grade 2-3


Additional criteria (two are required):


1. Anemia and a ≥2 mg/ml decrease from baseline hemoglobin level


2. A leukoerythroblastic peripheral blood picture


3. Increasing splenomegaly defined as either an increase in palpable splenomegaly of ≥5 cm


(distance of the tip of the spleen from the left costal margin) or the appearance of a newly


palpable splenomegaly


4. Increased lactate dehydrogenase (LDH) (above reference level)


5. Development of ≥1 of three constitutional symptoms: >10% weight loss in 6 months, night


sweats, unexplained fever (>37.5° C.)









The term “polycythemia vera” (PV), as used herein, is defined with reference to “The 2016 revision to the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia”, as published in Blood, 2016, 127:2391-2405. The term “post-polycythemia myelofibrosis” (PPV-MF), as used herein, refers to MF secondary to PV (i.e. MF arising as a progression of PV). According to the IWG-MRT criteria (Barosi G et al, Leukemia (2008) 22, 437-438), criteria for diagnosing post-polycythemia myelofibrosis are:









TABLE D





Criteria for diagnosis of post-polycythemia myelofibrosis















Required criteria:


1. Documentation of a previous diagnosis of polycythemia vera as defined by the WHO


criteria


2. Bone marrow fibrosis grade 2-3 (on 0-3 scale)


Additional criteria (two are required):


1. Anemia or sustained loss of requirement of either phlebotomy (in the absence of


cytoreductive therapy) or cytoreductive treatment for erythrocytosis


2. A leukoerythroblastic peripheral blood picture


3. Increasing splenomegaly defined as either an increase in palpable splenomegaly of ≥5 cm


(distance of the tip of the spleen from the left costal margin) or the appearance of a newly


palpable splenomegaly


4. Development of ≥1 of three constitutional symptoms: >10% weight loss in 6 months, night


sweats, unexplained fever (>37.5° C.)









As used herein, the following response criteria as defined by the International Working Group-Myeloproliferative Neoplasms Research and Treatment (IWG-MRT) and the European Leukemia Net (ELN) response criteria for MF (Tefferi et al, Blood 2013 122:1395-1398, which is incorporated by reference in its entirety) are used herein:









TABLE E







International Working Group-Myeloproliferative Neoplasms Research and Treatment


(IWG-MRT) and the European Leukemia Net (ELN) response criteria for myelofibrosis








Response
Required criteria (for all response categories, benefit must last for ≥12


categories
weeks to qualify as a response)





Complete
Bone marrow: *Age-adjusted normocellularity; <5% blasts; ≤grade 1 MF and


response
Peripheral blood: Hemoglobin ≥100 g/L and <UNL; neutrophil count ≥1 ×


(CR)
109/L and <UNL;



Platelet count ≥100 × 109/L and <UNL; <2% immature myeloid cells and



Clinical: Resolution of disease symptoms; spleen and liver not palpable; no



evidence of EMH


Partial
Peripheral blood: Hemoglobin ≥100 g/L and <UNL; neutrophil count ≥1 × 109/L


response
and <UNL; platelet count ≥100 × 109/L and <UNL; <2% immature myeloid


(PR)
cells and



Clinical: Resolution of disease symptoms; spleen and liver not palpable; no



evidence of EMH or



Bone marrow: *Age-adjusted normocellularity; <5% blasts; ≤grade 1 MF, and



peripheral blood: Hemoglobin ≥85 but <100 g/L and <UNL; neutrophil count ≥1



× 109/L and <UNL; platelet count ≥50, but <100 × 109/L and <UNL; <2%



immature myeloid cells and



Clinical: Resolution of disease symptoms; spleen and liver not palpable; no



evidence of EMH


Clinical
The achievement of anemia, spleen or symptoms response without


improvement
progressive disease or increase in severity of anemia, thrombocytopenia, or


(CI)
neutropenia§


Anemia
Transfusion-independent patients: a ≥20 g/L increase in hemoglobin level


response
Transfusion-dependent patients: becoming transfusion-independent


Spleen
A baseline splenomegaly that is palpable at 5-10 cm, below the LCM,


response#
becomes not palpable** or



A baseline splenomegaly that is palpable at >10 cm, below the LCM,



decreases by ≥50%**



A baseline splenomegaly that is palpable at <5 cm, below the LCM, is not



eligible for spleen response



A spleen response requires confirmation by MRI or computed tomography



showing ≥35% spleen volume reduction


Symptoms
A ≥50% reduction in the MPN-SAF TSS††


response











    • EMH, extramedullary hematopoiesis (no evidence of EMH implies the absence of pathology- or imaging study-proven nonhepatosplenic EMH); LCM, left costal margin; UNL, upper normal limit.

    • * Baseline and posttreatment bone marrow slides are to be interpreted at one sitting by a central review process.

    • † Grading of MF is according to the European classification: Thiele et al. European consensus on grading bone marrow fibrosis and assessment of cellularity. Haematologica. 2005; 90:1128.

    • ‡ Immature myeloid cells constitute blasts+promyelocytes+myelocytes+metamyelocytes+nucleated red blood cells. In splenectomized patients, <5% immature myeloid cells is allowed.

    • § Increase in severity of anemia constitutes the occurrence of new transfusion dependency or a ≥20 g/L decrease in hemoglobin level from pretreatment baseline that lasts for at least 12 weeks. Increase in severity of thrombocytopenia or neutropenia is defined as a 2-grade decline, from pretreatment baseline, in platelet count or absolute neutrophil count, according to the Common Terminology Criteria for Adverse Events (CTCAE) version 4.0. In addition, assignment to Cl requires a minimum platelet count of ≥25 000×10(9)/L and absolute neutrophil count of ≥0.5×10(9)/L.

    • ∥ Applicable only to patients with baseline hemoglobin of <100 g/L. In patients not meeting the strict criteria for transfusion dependency at the time of treatment initiation, but have received transfusions within the previous month, the pre-transfusion hemoglobin level should be used as the baseline.

    • ¶ Transfusion dependency is defined as transfusions of at least 6 units of packed red blood cells (PRBC), in the 12 weeks prior to start of treatment initiation, for a hemoglobin level of <85 g/L, in the absence of bleeding or treatment-induced anemia. In addition, the most recent transfusion episode must have occurred in the 28 days prior to start of treatment initiation. Response in transfusion-dependent patients requires absence of any PRBC transfusions during any consecutive “rolling” 12-week interval during the treatment phase, capped by a hemoglobin level of >85 g/L.

    • # In splenectomized patients, palpable hepatomegaly is substituted with the same measurement strategy.

    • ** Spleen or liver responses must be confirmed by imaging studies where a ≥35% reduction in spleen volume, as assessed by MRI or CT, is required. Furthermore, a ≥35% volume reduction in the spleen or liver, by MRI or CT, constitutes a response regardless of what is reported with physical examination.

    • †† Symptoms are evaluated by the MPN-SAF TSS. The MPN-SAF TSS is assessed by the patients themselves and this includes fatigue, concentration, early satiety, inactivity, night sweats, itching, bone pain, abdominal discomfort, weight loss, and fevers. Scoring is from 0 (absent/as good as it can be) to 10 (worst imaginable/as bad as it can be) for each item. The MPN-SAF TSS is the summation of all the individual scores (0-100 scale). Symptoms response requires ≥50% reduction in the MPN-SAF TSS.





In one embodiment the present invention provides an ERK1/2 inhibitor, suitably Compound A, alone or in combination with a JAK inhibitor, suitably ruxolitinib or a pharmaceutically acceptable salt thereof, for use in the treatment of myelofibrosis, especially primary MF, wherein the patient achieves complete response to the treatment according to the criteria in Table.


In one embodiment the present invention provides an ERK1/2 inhibitor, suitably Compound A, alone or in combination with a JAK inhibitor, suitably ruxolitinib or a pharmaceutically acceptable salt thereof, for use in the treatment of myelofibrosis, especially primary MF, wherein the patient achieves partial response to the treatment according to the criteria in Table.


Among patients, myelofibrosis frequently causes shortened survival due to disease transformation to acute leukemia, progression without acute transformation, cardiovascular complications or thrombosis, infection or portal hypertension. It is one of the aims of the present invention to improve the median survival of myelofibrosis patients.


As used herein, the term “median survival time” refers to the time of diagnosis or from the time of initiation of treatment according to the present invention that half of the patients in a group of patients diagnosed with the disease are still alive compared to patients receiving best available treatment or compared to patients receiving placebo and wherein patients belong to the same risk group of myelofibrosis, for example as described by Gangat et al (J Clin Oncol. 2011 Feb. 1; 29(4):392-397), which is hereby incorporated by reference in its entirety.


Accordingly, in one embodiment the present invention provides an ERK1/2 inhibitor, suitably Compound A, alone or in combination with a JAK inhibitor, suitably ruxolitinib or a pharmaceutically acceptable salt thereof, for use in the treatment of myelofibrosis, especially primary MF, wherein median survival time is increased by at least 3 months in the group of high risk MF patients or by at least six months, preferably by at least 12 months in the group of medium risk MF patients.


The combinations and the methods of the invention may be used to treat a patient as described herein.


As used herein, the term “patient” refers to a human being. The combinations described herein are suitable for treating human patients having a disorder that can be treated by modulating (e.g., augmenting or inhibiting) an immune response.


The patient may be a patient suffering from myeloproliferative neoplasms (MPNs) such as myelofibrosis (MF), essential thrombocythemia (ET) and/or polycythemia vera (PV). For example, the patient may be suffering from PMF, PPV-MF, or PET-MF.


In certain embodiments, the patient, e.g. an adult patient, suffering from PMF, PPV-MF, or PET-MF.


In certain embodiments, the patient is a patient suffering from PMF, PPV-MF, or PET-MF and, in addition at baseline, shows one or more, or all characteristics selected from: (a) has Hb<11 g/dL (≤6.8 mmol/L); (b) is responsive and/or stable on treatment with a JAK inhibitor, such as ruxolitinib, and (c) exhibits measurable splenomegaly with spleen volume of ≥450 cm3 by MRI or CT scan or by palpable spleen measuring of ≥5 cm below left costal margin (LCM).


In certain embodiments, the human patient has a disorder described herein, e.g., myeloproliferative neoplasms (MPNs) such as myelofibrosis (MF), essential thrombocythemia (ET) and/or polycythemia vera (PV), is responsive and/or stable on treatment with a JAK inhibitor, such as ruxolitinib, and is in need for additional treatment options. In certain embodiments, the human patient has hemoglobin level less than 10 g/dL, a confirmed diagnosis of PMF, PPV-MF, or PET-MF, a palpable spleen of at least 5 cm from the left costal margin (LCM) and/or enlarged spleen volume of at least 450 cm3 per MRI or CT-scan, responsive and/or stable on JAK inhibitor therapy such as ruxolitinib and in need of additional treatment options. In certain embodiments, the human patients with PMF, PPV-MF, or PET-MF and receiving treatment with combinations described herein achieve a hemoglobin improvement of 2.0 g/dL or ≥1.5 g/dL from baseline, arrestment and/or improvement in spleen size, and/or improvement in bone marrow fibrosis of ≥1 grade from baseline.


The expression “is responsive and/or stable on treatment with a JAK inhibitor, such as ruxolitinib” means for example, being on ruxolitinib therapy for a period of time such as at least 12 weeks, with an unchanged ruxolitinib dose (e.g. in the range 5-25 mg BID) for the previous ≥4 weeks prior to first dose of treatment. It can also mean that such a patient is on ruxolitinib therapy for a period of time such as at least 24 weeks, with an unchanged ruxolitinib dose (e.g. in the range 5-25 mg BID) for the previous ≥8 weeks prior to first dose of treatment.


The term “treat”, “treating”, “treatment” or “therapy”, as used herein, means obtaining beneficial or desired results, for example, clinical results. Beneficial or desired results can include, but are not limited to, alleviation of one or more symptoms, as defined herein. One aspect of the treatment is, for example, that said treatment should have a minimal adverse effect on the patient, e.g. the agent used should have a high level of safety, for example without producing the side effects of a previously known therapy. The term “alleviation”, for example in reference to a symptom of a condition, as used herein, refers to reducing at least one of the frequency and amplitude of a symptom of a condition in a patient.


As used herein, the term “newly diagnosed” refers to diagnosis of the disorder, e.g. myelofibrosis and said patient has not received any treatment for the disorder. In one embodiment, the present invention provides an ERK1/2 inhibitor (e.g. Compound A) or a pharmaceutical acceptable salt thereof, alone or in combination with a JAK inhibitor, suitably ruxolitinib or a pharmaceutically acceptable salt thereof, for use in the treatment of a newly diagnosed myelofibrosis patient.


The term “triple-negative myelofibrosis patient”, as used herein, refers to a patient who lacks JAK2, CALR and MPL mutations. In one embodiment the present invention provides an ERK1/2 inhibitor (e.g. Compound A) or a pharmaceutical acceptable salt thereof, alone or in combination with a JAK inhibitor, suitably ruxolitinib or a pharmaceutically acceptable salt thereof, for use in the treatment of triple-negative myelofibrosis patient.


The term “best available therapy”, as used herein, refers to any commercially available agent approved, for example prior to March 2018, or prior to October 2020, for the treatment of PMF, PET-MF or PPV-MF, as monotherapy, or in combination. Exemplary agents include, but are not limited to ruxolitinib or a pharmaceutically acceptable salt thereof, antineoplastic agents (e.g., hydroxyurea, anagrelide), glucocorticoids (e.g., prednisone/prednisolone, methylprednisolone), antianemia preparations (e.g., epoetin-alpha), immunomodulatory agents (e.g., thalidomide, lenalidomide), purine analogs (e.g., mercaptopurine, thioguanine), antigonadotropins (e.g., danazol), interferons (e.g., PEG-interferon-alpha 2a, interferon-alpha), nitrogen mustard analogs (e.g. melphalan), pyrimidine analogs (e.g., cytarabine).


The term “splenomegaly”, as used herein, refers to a palpably enlarged spleen (e.g. a spleen is palpable at ≥5 cm below the left coastal margin) or to an enlarged spleen as detected by an imaging test (e.g. a computed tomography (CT) scan, MRI, X-rays or ultrasound), wherein the term “enlarged spleen” refers to a spleen greater in size than normal (e.g., median normal spleen volume of 200 cm3).


The term “treatment of splenomegaly”, as used herein, refers to “improvement of splenomegaly”, which means a decrease in splenomegaly, for example a reduction in spleen volume, as defined by the International Working Group-Myeloproliferative Neoplasms Research and Treatment (IWG-MRT) and the European Leukemia Net (ELN) response criteria for MF in Table. In one embodiment, the invention may provide the use of an ERK1/2 inhibitor (e.g. Compound A) or a pharmaceutical acceptable salt thereof, alone or in combination with ruxolitinib or a pharmaceutically acceptable salt thereof for treatment of myelofibrosis, particularly for the treatment of splenomegaly associated with myelofibrosis, resulting in, for example, ≥20%, ≥25%, ≥30% or ≥35% reduction in spleen volume as measured by magnetic resonance imaging (MRI) or computed tomography (CT) from pre-treatment baseline to, for example, week 24 or week 48.


The term “hepatomegaly”, as used herein, refers to a palpably enlarged liver or to an enlarged liver as detected by an imaging test (e.g. a computed tomography (CT) scan), wherein the term “enlarged liver” refers to a liver greater in size than normal (e.g., median normal liver volume of approximately 1500 cm3).


The term “treatment of hepatomegaly”, as used herein, refers to “improvement of hepatomegaly”, which means a decrease in hepatomegaly, for example a reduction in hepatomegaly, as defined according to the International Working Group-Myeloproliferative Neoplasms Research and Treatment (IWG-MRT) and the European Leukemia Net (ELN) response criteria for MF in the preceding table. Accordingly, in one embodiment, the present invention provides the use of an ERK1/2 inhibitor (e.g. Compound A) or a pharmaceutical acceptable salt thereof, alone or in combination with ruxolitinib or a pharmaceutically acceptable salt thereof for treatment of myelofibrosis, particularly for the treatment of hepatomegaly associated with myelofibrosis, resulting in, for example, ≥20%, ≥25%, ≥30% or ≥35% reduction in liver volume as measured by magnetic resonance imaging (MRI) or computed tomography (CT) from pre-treatment baseline to, for example, week 24 or week 48.


The term “thrombocytopenia”, as used herein, refers to a platelet count, in blood specimen laboratory test, lower than normal, or less than 150,000/ml. The term “severity of thrombocytopenia”, as used herein, refers, for example, to specific grade 1-4 of thrombocytopenia according to CTCAE (version 4.03).


The term “treatment of thrombocytopenia”, as used herein, refers to “stabilizing thrombocytopenia” or “improving thrombocytopenia”, in comparison to the pre-treatment situation or in comparison to best available therapy or to placebo control. The term “stabilizing thrombocytopenia” refers, for example, to prevent an increase in the severity of thrombocytopenia, namely the platelet count remains stable. The term “improving thrombocytopenia” refers to alleviation of the severity of thrombocytopenia, namely increasing blood platelet count. In one embodiment, the invention provides an ERK1/2 inhibitor (e.g. Compound A) or a pharmaceutical acceptable salt thereof, alone or in combination with ruxolitinib or a pharmaceutically acceptable salt thereof, for use in the treatment of myelofibrosis, particularly for the treatment of thrombocytopenia associated with myelofibrosis, resulting in stabilizing thrombocytopenia or improving thrombocytopenia from pre-treatment baseline to, for example, week 24 or week 48 of treatment.


The term “neutropenia”, as used herein, refers to an absolute neutrophil count (ANC), in blood specimen laboratory test, lower than normal value, or less than 1500/ml. The term “severity of neutropenia”, as used herein, refers, for example, to specific grade 1-4 of neutropenia according to CTCAE (version 4.03).


The term “treatment of neutropenia”, as used herein, refers to “stabilizing neutropenia” or “improving neutropenia”, for example, in comparison to the pre-treatment situation or in comparison to best available therapy or to placebo control. The term “stabilizing neutropenia” refers, for example, to prevent an increase in the severity of neutropenia. The term “improving neutropenia” refers, for example, to a decrease in the severity of neutropenia. In one embodiment, the invention provides an ERK1/2 inhibitor (e.g. Compound A) or a pharmaceutical acceptable salt thereof, alone or in combination with ruxolitinib or a pharmaceutically acceptable salt thereof, for use in the treatment of myelofibrosis, particularly for the treatment of neutropenia associated with myelofibrosis, resulting in stabilizing neutropenia or improving neutropenia from pre-treatment baseline to, for example, week 24 or week 48 of treatment.


The term “anemia”, as used herein, refers to hemoglobin level, in blood specimen laboratory test, of less than 13.5 gram/100 ml in men and hemoglobin level of less than 12.0 gram/100 ml in women. The term “severity of anemia”, as used herein, refers, for example, to specific grade 1-4 of anemia according to CTCAE (version 4.03)].


The term “treatment of anemia”, as used herein, refers to “stabilizing anemia” or “improving anemia”, for example, in comparison to the pre-treatment situation or in comparison to best available therapy or to placebo control. The term “stabilizing anemia” refers, for example, to prevent an increase in the severity of anemia (e.g. preventing that a “transfusion-independent” patient becomes a “transfusion-dependent” patient or preventing anemia grade 2 becomes anemia grade 3). The term “improving anemia” refers to a decrease in the severity of anemia or an improvement in hemoglobin level. In one embodiment, the invention may provide the use of an ERK1/2 inhibitor (e.g. Compound A) or a pharmaceutical acceptable salt thereof, alone or in combination with ruxolitinib or a pharmaceutically acceptable salt thereof, for treatment of myelofibrosis, particularly for the treatment of anemia associated with myelofibrosis, resulting in stabilizing anemia or improving anemia from pre-treatment baseline to, for example, week 24 or week 48 of treatment.


The term “treatment of bone marrow fibrosis associated with MF”, as used herein, means “stabilizing bone marrow fibrosis” or “improving bone marrow fibrosis”, for example, in comparison to the pre-treatment situation or in comparison to best available therapy or to placebo control. The term “stabilizing bone marrow fibrosis” refers, for example, to prevent increase in severity of bone marrow fibrosis. The term “improving bone marrow fibrosis” refers to a decrease in severity of bone marrow fibrosis, for example, from pre-treatment baseline, according to the 2005 European consensus grading system. In one embodiment, the invention may provide the use of an ERK1/2 inhibitor (e.g. Compound A) or a pharmaceutical acceptable salt thereof, alone or in combination with ruxolitinib or a pharmaceutically acceptable salt thereof, for treatment of myelofibrosis, particularly for the treatment of bone marrow fibrosis associated with MF, resulting in stabilizing bone marrow fibrosis or improving bone marrow fibrosis from pre-treatment baseline to, for example, week 24 or week 48 of treatment.


The term “constitutional symptoms associated with myelofibrosis”, as used herein, refers to common debilitating chronic myelofibrosis symptoms, such as fever, pruritus (i.e. itching), abdominal pain/discomfort, weight loss, fatigue, inactivity, early satiety, night sweats or bone pain; for example, as described by Mughal et al (Int J Gen Med. 2014 Jan. 29; 7:89-101).


The term “treatment of constitutional symptoms associated with myelofibrosis”, as used herein, refers to “improvement of constitutional symptoms associated with myelofibrosis”, for example, in comparison to the pre-treatment situation or in comparison to best available therapy or to placebo control, for example, a reduction in total symptom score as measured by the modified myelofibrosis symptom assessment form version 2.0 diary (modified MFSAF v2.0) (Cancer 2011; 117:4869-77; N Engl J Med 2012; 366:799-807, the entire contents of which are incorporated herein by reference). In one embodiment, the invention may provide the use of an ERK1/2 inhibitor (e.g. Compound A) or a pharmaceutical acceptable salt thereof, alone or in combination with ruxolitinib or a pharmaceutically acceptable salt thereof, for treatment of myelofibrosis, particularly for the treatment of constitutional symptoms associated with myelofibrosis, resulting in improvement of constitutional symptoms associated with myelofibrosis from pre-treatment baseline to, for example, week 24 or week 48 of treatment.


In another embodiment of any use of the invention, one or more of the constitutional symptoms associated with MF are alleviated (e.g. by eliminating or by reducing intensity, duration or frequency). In one embodiment, the reduction of constitutional symptoms is at least ≥20%, at least ≥30%, at least ≥40% or at least ≥50% as assessed by the modified MFSAF v2.0 from pre-treatment baseline to, for example, week 24 or week 48.


In one embodiment of any use of the invention, the ERK1/2 inhibitor, suitably Compound A, is administered subsequently or prior to splenectomy or radiotherapy, such as splenic irradiation.


Combination Therapy

In one aspect the present invention provides an ERK1/2 inhibitor, suitably Compound A, for use in the treatment of MF, wherein the ERK1/2 inhibitor is administered in combination with at least one further active agent.


In one embodiment the at least one agent is an inhibitor of a non-receptor tyrosine kinases, the Janus kinases (JAK). A considerable number of cytokine and growth factor receptors utilize non-receptor tyrosine kinases, the Janus kinases (JAK), to transmit extracellular ligand binding into an intracellular response. For example, erythropoietin, thrombopoietin and granulocyte monocyte colony stimulating factor are all known to signal through receptors that utilize JAK2. JAK activate a number of downstream pathways implicated in proliferation and survival, including the STATs (signal transducers and activators of transcription), a family of important latent transcription factors.


Accordingly, the present invention relates to the combination use of an ERK1/2 inhibitor (e.g., Compound A) or a pharmaceutical acceptable salt thereof, with at least one JAK inhibitor, suitably ruxolitinib or a pharmaceutically acceptable salt thereof.


In one embodiment the at least one further active agent is a JAK1/JAK2 inhibitor, suitably ruxolitinib, or a pharmaceutically acceptable salt thereof, or momelotinib, or a pharmaceutically acceptable salt thereof; more suitably ruxolitinib or a pharmaceutically acceptable salt, more suitably ruxolitinib phosphate.


In one embodiment the at least one further active agent is a JAK2/FLT3 inhibitor, suitably pacritinib or a pharmaceutically acceptable salt thereof or fedratinib or a pharmaceutically acceptable salt thereof.


In one embodiment the at least one further active agent is a JAK2V617F inhibitor, suitably gandotinib or a pharmaceutically acceptable salt thereof.


In one embodiment the at least one further active agent is a JAK2 inhibitor, suitably BMS-911543 or a pharmaceutically acceptable salt thereof.


In one embodiment the at least one further active agent is a JAK1 inhibitor, suitably itacitinib or a pharmaceutically acceptable salt thereof, in particular itacitinib adipate.


In one embodiment the at least one further active agent is a JAK2/Src inhibitor, suitably NS-018 or a pharmaceutically acceptable salt thereof.


The present invention provides a pharmaceutical combination comprising a JAK 1/2 inhibitor such as ruxolitinib, or a pharmaceutically acceptable salt thereof, and an ERK 1/2 inhibitor such as Compound A, or a pharmaceutically acceptable salt thereof.


In one aspect, the present invention provides a pharmaceutical combination comprising, consisting essentially of, or consisting of Compound A, or a pharmaceutical acceptable salt thereof, and b) a JAK1/2 inhibitor, suitably ruxolitinib, or a pharmaceutically acceptable salt thereof. Suitably the pharmaceutical combination is for use in the treatment of myelofibrosis.


In one aspect, the present invention provides Compound A, or a pharmaceutical acceptable salt thereof, for use in the treatment of myelofibrosis, wherein Compound A, or a pharmaceutical acceptable salt thereof, is administered in combination with ruxolitinib, or a pharmaceutically acceptable salt thereof, and wherein Compound A, or a pharmaceutical acceptable salt thereof, and ruxolitinib, or a pharmaceutically acceptable salt thereof, are administered in jointly therapeutically effective amounts.


In one aspect, the present invention provides ruxolitinib, or a pharmaceutically acceptable salt thereof, for use in the treatment of myelofibrosis, wherein ruxolitinib, or a pharmaceutically acceptable salt thereof, is administered in combination with Compound A, or a pharmaceutical acceptable salt thereof, and wherein ruxolitinib, or a pharmaceutically acceptable salt thereof, and Compound A, or a pharmaceutical acceptable salt thereof, are administered in jointly therapeutically effective amounts.


The term “combination” or “pharmaceutical combination” used herein, refers to a non-fixed combination where an active agent and at least one further active agent may be administered independently at the same time or separately within time intervals, especially where these time intervals allow that the combination partners show a cooperative, e.g. synergistic effect. The terms “co-administration” or “combined administration” or the like as utilized herein are meant to encompass administration of the selected combination partner to a single patient in need thereof (e.g. a patient), and are intended to include treatment regimens in which the agents are not necessarily administered by the same route of administration or at the same time.


The term “non-fixed combination” means that the active ingredients, e.g. one active agent and at least one further active agent, are both administered to a patient as separate entities either simultaneously or sequentially with no specific time limits, wherein such administration provides therapeutically effective levels of the two compounds in the body of the patient. In particular, reference to an ERK1/2 inhibitor or a pharmaceutical acceptable salt thereof in combination with ruxolitinib or a pharmaceutically acceptable salt thereof as used herein (e.g. in any of the embodiments or in any of the claims herein), refers to a “non-fixed combination”; and reference to ruxolitinib or a pharmaceutically acceptable salt thereof as used herein (e.g. in any of the embodiments or in any of the claims herein), in combination with at least one further active agent (Compound A being excluded) refers to either a fixed combination in one unit dosage form (e.g., capsule, tablet, caplets or particulates), a non-fixed combination, or a kit-of-parts for the combined administration wherein ruxolitinib or a pharmaceutically acceptable salt thereof and one or more combination partner (e.g. another drug as specified herein, also referred to as further “pharmaceutical active ingredient”, “therapeutic agent” or “co-agent”) may be administered independently at the same time or separately within time intervals.


A “pharmaceutical combination” as described herein preferably refers to a pharmaceutical combination comprising ruxolitinib, or a pharmaceutically acceptable salt thereof, and an ERK 1/2 inhibitor such as Compound A, or a pharmaceutically acceptable salt thereof.


The term “therapeutically effective amount” refers to an amount of a drug or a therapeutic agent that will elicit the desired biological and/or medical response of a tissue, system or an animal (including man) that is being sought by a researcher or clinician.


Administration and Treatment Regimen

A pharmacokinetic drug-drug interaction (PK DDI) between ruxolitinib and Compound A is unlikely or predicted to be low, based on a physiologically based pharmacokinetic (PBPK) model (SimCyp) analysis.


Dosing for Compound A (at ≤300 mg QD) and ruxolitinib (between 5 and 25 mg BID), a minimal DDI effect is anticipated with a predicted <1.3-fold transient increase of ruxolitinib exposure (AUC and Cmax), through CYP3A4 inhibition. This transient and limited increase in the exposure of ruxolitinib is unlikely to necessitate a ruxolitinib dose adjustment during the period of co-administration with Compound A. No change of Compound A systemic exposure is expected in the presence of ruxolitinib.


The present invention thus provides for dosages and administration regimens as follows.


Compound A may be administered either QD (once a day) or BID (twice a day), preferably QD. Preferably, the total daily dose (TTD) of Compound A is from 100-300 mg, or from 150-200 mg, or from 200-300 mg, e.g may be selected from 50, 100, 150, 200, 250 and 300 mg, preferably administered QD.


In one embodiment, Compound A, or a pharmaceutically acceptable salt thereof. may be administered orally at a daily dose of 100 mg, 200 mg, or 300 mg. preferably once daily.


Compound A can be generally administered in a unit dosage of about 1-2000 mg of active ingredient for a patient of about 50-70 kg, or about 1-500 mg or about 1-250 mg or about 1-150 mg or about 0.5-100 mg, or about 1-50 mg of active ingredient. The unit dosage may be administered once or repeatedly during the same day, or during the week. More specifically, a daily dose of between 45 mg and 600 mg, or between 100 mg and 450 mg, particularly between 150 mg and 300 mg, or between 200 mg and 300 mg may be suitable.


In one embodiment, Compound A is prepared for administration via oral delivery, and may be used as its hydrochloride salt. In some embodiments, the compound or its HCl salt is simply encapsulated in a pharmaceutically acceptable container such as a hard or soft gelcap for oral administration.


In one embodiment, the present invention provides the ERK1/2 inhibitor such as Compound A or a pharmaceutical acceptable salt thereof, for use in the treatment of myelofibrosis, wherein said ERK1/2 inhibitor, is administered in combination with ruxolitinib, or a pharmaceutically acceptable salt thereof. Suitably, ruxolitinib is administered in an amount of from 5 mg twice daily to 25 mg twice daily, such as 5 mg twice daily, 10 mg twice daily, 15 mg twice daily, 20 mg twice daily or 25 mg twice daily, depending on the patient's blood count according to the prescribing information for Jakavi®/Jakafi® and the judgment of the treating physician.


All publications and patent applications mentioned in the specification are indicative of the level of skill of those skilled in the art to which the presently disclosed inventive concepts pertain. All publications and patent applications are herein incorporated by reference to the same extent as if each individual publication or patent application was specifically and individually indicated to be incorporated by reference.


References in this specification to “the invention” are intended to reflect embodiments of the several inventions disclosed in this specification, and should not be taken as unnecessarily limiting of the claimed subject matter.


LIST OF ABBREVIATIONS





    • AE adverse event

    • AML Acute myeloid leukemia

    • ANC Absolute neutrophil count

    • ASCT allogeneic hematopoietic stem cell transplantation

    • AUC Area under curve

    • BID twice a day

    • BM bone marrow

    • C1D1 Cycle 1 Day 1 (and sequentially for other cycles and days, eg C1 D2, C2D1 etc.)

    • CT computed tomography

    • CTCAE Common Terminology Criteria for Adverse Events

    • CYP cytochrome P-450

    • DDI Drug-drug interaction

    • DLT dose-limiting toxicity

    • ECG Electrocardiogram

    • EORTC European Organization for Research and Treatment of Cancer

    • ERK extracellular signal-regulated kinase

    • ET essential thrombocythemia

    • Hb hemoglobin

    • IV Intravenous

    • IWG-MRT International Working Group-Myeloproliferative Neoplasms Research and Treatment

    • JAK Janus kinase

    • LCM left costal margin

    • MF myelofibrosis

    • MPN myeloproliferative neoplasm

    • MRI magnetic resonance imaging

    • PD pharmacodynamic(s)

    • PFS progression free survival

    • PK pharmacokinetic(s)

    • PLT platelets

    • PMF primary myelofibrosis

    • PRBC packed red blood cells

    • PV polycythemia vera

    • Q4W every 4 weeks

    • QD once a day

    • QLQ-C30 Quality of Life Questionnaire-Core 30

    • QoL quality of life

    • RBC red blood cell(s)

    • RP2D recommended phase 2 dose

    • RR Response rate

    • SAF symptom assessment form

    • STAT signal transducer and activator of transcription

    • TLS Tumor lysis syndrome

    • TSS total symptom score

    • WHO World Health Organization





The Examples below are set forth to aid in the understanding of the inventions but are not intended to, and should not be construed to, limit its scope in any way.


EXAMPLES
Example 1: Compound a (LTT462) in Combination with Ruxolitinib in MPN Cell Lines

The antiproliferative activity of Compound A (LTT462), Ruxolitinib, and ruxolitinib and Compound A was tested in the human line SET2 carrying the Jak2V617F mutation and in the murine Ba/F3 cell line stably expressing erythropoietin receptor (EpoR) as well as either wild-type JAK2 or Jak2V617F. To assess anti-proliferative effects of inhibitors, cells were seeded at 10′000/200 ul with increasing inhibitor concentrations in triplicate. Proliferation was assessed at 48 h using the CellTiter-Glo viability assay (Promega) and normalized to cell growth in medium with equivalent volume of DMSO. The concentration inhibiting proliferation by 50% (IC50) was determined with GraphPad Prism 8.0.


Combined JAK2 inhibition by ruxolitinib and ERK1/2 inhibition by Compound A was able to reduce the IC50 in Ba/F3 JAK2V617F cells (FIG. 1A) by 3-fold as compared to ruxolitinib as a single agent, while there was less benefit of Compound A in addition to ruxolitinib in SET2 cells (FIG. 1B). In Ba/F3 EpoR cells with wild-type Jak2, the addition of Compound A to ruxolitinib did not increase efficacy as much as in Jak2 V617F mutant cells.


Example 2: Compound a (LTT462) in Combination with Ruxolitinib in MPN Mouse Models

The antiproliferative activity of Compound A (LTT462), Ruxolitinib, and ruxolitinib and Compound A was tested using a Jak2V617F knock-in mouse model. Primarily, a Jak2V617F knock-in mouse model reflecting a polycythemia vera phenotype was used (Mullally A et al, Cancer Cell 2010), which is characterized by Jak2V617F expression in hematopoietic tissues based on expression of Cre-recombinase under the control of the Vav or the Mx-1 promoter.


For treatment studies, bone marrow (BM) from primary Jak2V617F Vav-Cre CD45.2 mice was mixed 1:1 with Jak2 wild-type CD45.1 BM and transplanted into lethally irradiated CD45.1 recipients. Development of the MPN phenotype was confirmed by peripheral blood counts 2 months after BM transplantation. Mice were randomized to treatment groups according to blood counts and treated by oral gavage for 1-4 weeks.


As a model of MPLW515L mutant MPN, CD117-enriched (Miltenyi) Balb/c BM was transduced with retroviral supernatant containing MSCV-hMPLW515L-IRES-GFP and injected i.v. into lethally irradiated Balb/c recipients. Development of the MPN phenotype was confirmed by blood counts 2-4 weeks after BM transplantation. For treatment studies, mice were randomized to treatment groups according to blood counts and treated by oral gavage for 1-4 weeks. The suppression of ERK signaling was confirmed in splenocytes by Western blot analysis. For histopathology, tissues were fixed in 4% paraformaldehyde, paraffin-embedded and stained with hematoxylin/eosin. Gomori stain was used for assessment of reticuline fibers. Fibrosis was graded by a specialized hematopathologist. For flow cytometry analyses, BM cells were stained for lineage markers, Sca-1, c-Kit, CD41, CD150, CD48, CD16/32 and CD105, CD71 and Ter-119 (eBioscience) and for CD45.1 and CD45.2 alleles to assess mutant allele burden as the fraction of CD45.2+ total BM or erythroid progenitor cells. Analyses were performed on a LSRFortessa (BD).


Combined JAK2 and ERK1/2 inhibition with ruxolitinib and Compound A inhibited activation of ERK downstream targets RSK3 and DUSP6 in the splenocytes of Jak2V617F mice (FIG. 2A) and potently corrected splenomegaly (FIG. 2C) and polyglobulia or polycythemia (FIG. 2B). In MPLW515L mice, combined JAK2 and ERK1/2 inhibition corrected leukocytosis and reduced splenomegaly and bone marrow fibrosis to an extent which was superior to single agent therapies (FIG. 2D).


Example 3: Dose Response of Compound a (LTT462) in Combination with Ruxolitinib in JAK2VF Mouse Model

Compound A in combination with ruxolitinib significantly normalized splenomegaly, polycythemia and hematocrit in JAK2V617F PV/MF mouse model.


Mice were administered an oral dose of either vehicle, ruxolitinib at 60 mg/kg BID, Compound A at 75 mg/kg QD, or the combination of ruxolitinib at 60 mg/kg BID and Compound A at 75 mg/kg QD for 14 consecutive days.


Combined treatment with ruxolitinib and Compound A significantly reduced elevated hematocrit (FIG. 3A) and splenomegaly (FIG. 3B) as compared to either single agent and vehicle-treated mice.


Furthermore, the tested combination was well tolerated as judged by lack of body weight loss (FIG. 4A), bone marrow cellularity (FIG. 4B), normal WBC count (FIG. 4C), and normal platelet count (FIG. 4D).


These results support the potential exploration of the combination of ruxolitinib and Compound A in polycythemia vera and myelofibrosis.


Example 4: Compound a (LTT462) in Combination with Ruxolitinib in MPN Cells, JAK2V617F and MPLW5151L Mutant Mouse Models

Compound a (LTT462) in Combination with Ruxolitinib in CD34+ Peripheral Blood Mononuclear Cells (PBMCs)


Combined treatment with ruxolitinib and Compound A suppressed growth of the colonies derived from CD34+ PBMCs of a MF patient more potently than Compound A or ruxolitinib alone (FIG. 5A).


Compound a (LTT462) in Combination with Ruxolitinib in Jak2V617F Mice


Combined treatment with ruxolitinib and Compound A resulted in superior reduction of hematocrit than Compound A or ruxolitinib alone (FIG. 5B).


Compound a (LTT462) in Combination with Ruxolitinib in EpoR Jak2V617F Mutant and Jak2 Wild Type Ba/F3 Cells


As seen by a stronger decrease in IC50 value, combined treatment with ruxolitinib and Compound A further suppressed proliferation of EpoR Jak2V617F mutant and Jak2 wild type Ba/F3 cells as compared to ruxolitinib or Compound A alone (FIG. 6).


These results support the potential exploration of the combination of ruxolitinib and Compound A in myeloproliferative neoplasms.


Example 5: Genetic Deletion of ERK1/2 in MPN Mouse Model

Jak2V617F Mx-Cre mouse was crossed with Erk1−/− Erk2 fl/fl Mx-Cre mice to genetically delete ERK1/2 to obtain Jak2V617F Erk1−/− Erk2 fl/fl Mx-Cre offspring. BM from primary Jak2V617F Mx-Cre CD45.2 or Jak2V617F Erk1−/− Erk2 fl/fl Mx-Cre CD45.2 mice was mixed 1:1 with Jak2 WT 45.1 BM and injected into lethally irradiated CD45.1 mice for treatment studies. Recipient mice were treated with 5 doses of poly I:C (300 ug/dose) five weeks after transplantation to induce ERK1/2 loss, were randomized according to blood counts and treated with ruxolitinib for 2 weeks. ERK1/2 deletion combined with Jak2 inhibition with ruxolitinib enhanced therapeutic efficacy of either JAK2 inhibition or ERK1/2 deletion as single interventions with pronounced reductions of the MPN clone and correction of the MPN phenotype.


Combined ruxolitinib and ERK1/2 deletion are more effective in reducing the mutant clone (FIG. 7A) and normalizing red blood cell count (7B) in a Jak2V617F mouse model of MPN.


Example 6: A Randomized, Open-Label, Phase 1/11 Open Platform Study Evaluating Safety and Efficacy of a Combination of Ruxolitinib and Compound a in Myelofibrosis Patients

The efficacy of the combination of ruxolitinib and Compound A may be evaluated as follows.


The purpose of this study is to investigate the safety, pharmacokinetics and efficacy of combination treatment of ruxolitinib with Compound A in MF patients. The study consists of three parts:

    • Part 1: Dose escalation and safety run-in (recommended Phase II dose confirmation)
    • Part 2: Selection
    • Part 3: Expansion


Myelofibrosis (MF) is defined by progressive bone marrow (BM) fibrosis and a consecutive reduction of blood cells. The disruption of the medullary erythropoietic niche is the primary mechanism governing the bone marrow failure and anemia, which typify MF. Nearly 40% of MF patients have hemoglobin (Hb) levels <10 g/dL at diagnosis. Furthermore, anemia is the disease feature most consistently associated with poor prognosis in MF.


This combination therapy may deliver transformational clinical benefits such as improvement of progression free survival (PFS) as a consequence of superior disease control or reduction of the malignant clone, associated with an improvement of cytopenia and in particular anemia, as well as improvement in quality of life (QoL) as captured by relevant patient reported outcomes measurements (PROs).


Key Inclusion Criteria:

    • Patients have diagnosis of primary myelofibrosis (PMF) according to the 2016 World Health Organization (WHO) criteria, or diagnosis of postessential thrombocythemia (ET) (PET-MF) or post-polycythemia vera (PV) myelofibrosis (PPV-MF) according to the International Working Group for Myelofibrosis Research and Treatment (IWG-MRT) 2007 criteria;
    • Palpable spleen of at least 5 cm from the left costal margin (LCM) to the point of greatest splenic protrusion or enlarged spleen volume of at least 450 cm3 per MRI or CT scan at baseline (a MRI/CT scan up to 8 weeks prior to first dose of study treatment can be accepted).


Have been treated with ruxolitinib for at least 24 weeks prior to first dose of study treatment.


Are stable (no dose adjustments) on the prescribed ruxolitinib dose (between 5 and 25 mg twice a day (BID)) for 8 weeks prior to first dose of study treatment.

    • Hemoglobin <10 g/dL
    • Part 1: Platelet counts ≥75 000/μL
    • Part 2: Platelet counts ≥50 000/μL.


Key Exclusion Criteria


Patients will not be eligible for inclusion in the study if they meet any of the following criteria:


Not able to understand and to comply with study instructions and requirements.


Received any investigational agent for the treatment of MF (except ruxolitinib) within 30 days of first dose of study treatment or within 5 halflives of the study treatment, whichever is greater.


Peripheral blood blasts count of >10%.


Received a monoclonal antibody (Ab) or immunoglobulin-based agent within 1 year of screening, or has documented severe hypersensitivity reactions/immunogenicity (IG) to a prior biologic.


Splenic irradiation within 6 months prior to the first dose of study drug.


Received blood platelet transfusion within 28 days prior to first dose of study treatment.


Patients with known TP53 mutation or deletion of TP53.


Primary Objectives:


The primary objective of Part 1 of this study is to characterize the safety, tolerability, and the recommended Phase 2 dose (RP2D) of each combination partner used with ruxolitinib in subjects with myelofibrosis. This may be assessed by the incidence and severity of dose-limiting toxicity (DLTs) within the first 2 treatment cycles in Part 1 of the study.


The primary objective of Parts 2 and 3 of the study is to evaluate the preliminary efficacy of the novel ruxolitinib combination treatments in subjects with myelofibrosis. This may be based on the assessment of the response rate (RR) at the end of Week 24 or Cycle 6. The RR is the composite of anemia improvement of ≥1.5 g/dL, no spleen volume progression and no symptom worsening.


Primary Endpoints:


Response rate (RR) for the composite endpoint (anemia improvement of ≥1.5 g/dL and no spleen volume progression and no symptom worsening) at the end of Cycle 6.


Incidence and severity of dose limiting toxicity (DLT) within the first 2 treatment cycles in Part 1 of the study.


Secondary Objectives:


To assess the proportion of patients in each treatment arm who achieved an Hb improvement of ≥2.0 g/dL or ≥1.5 g/dL (Parts 2 & 3).


To evaluate changes in symptoms of myelofibrosis in each treatment arm using MFSAF v4.0 and European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire-Core 30 (QLQ-C30) patient reported outcomes (PROs) from baseline (Parts 2 & 3).


To characterize the pharmacokinetic profile of ruxolitinib administered in combination with Compound A (Parts 1, 2 & 3).


To evaluate the changes in spleen size in each treatment arm (Parts 2 & 3).


To evaluate the effect of ruxolitinib combination treatment in delaying progression of MF and estimate time to progression free survival (PFS) event (Parts 2 & 3).


To evaluate the effect on bone marrow fibrosis in each treatment arm by determining the proportion of subjects achieving improvement in bone marrow fibrosis of ≥1 grade from baseline (Parts 2 & 3).


To evaluate long-term safety and tolerability of ruxolitinib and Compound A combination treatment (Parts 1, 2 & 3).


Secondary Endpoints:


Change in MFSAF v4.0 and EORTC QLQ-C30 from Baseline.


PK parameters (e.g., AUC, Cmax, Tmax) and concentration vs. time profiles of each investigational drug within combination regimens.


Change in spleen length (by palpation) from baseline.


Change in spleen volume (by MRI/CT) from baseline.


Estimate of progression free survival (PFS) where events are defined as follows:

    • Progressive splenomegaly as assessed by increasing spleen volume (by MRI/CT) of ≥25% from baseline. The progression date will be the date of MRI/CT assessment confirming spleen volume increase of ≥25% from baseline;
    • Accelerated phase defined by a circulating peripheral blood blast content of >10% but <20% confirmed after 2 weeks. The progression date will be the date of first increase in peripheral blood blast content of >10%;
    • Deteriorating cytopenia (dCP) independent from treatment defined for all patients by platelet count <35×10{circumflex over ( )}9/L or neutrophil count <0.75×10{circumflex over ( )}9/L that lasts for at least 4 weeks. The progression date will be the date of first decrease of platelets <35×10{circumflex over ( )}9/L or neutrophils <0.75×10{circumflex over ( )}9/L confirmed after 4 weeks;
    • Leukemic transformation defined by a peripheral blood blast content of ≥20% associated with an absolute blast count of ≥1×10{circumflex over ( )}9/L that lasts for at least 2 weeks or a bone marrow blast count of ≥20%. The progression date will be the date of first increase in peripheral blood blast content of ≥20% associated with an absolute blast count of ≥1×10{circumflex over ( )}9/L OR the date of the bone marrow blast count of ≥20%;
    • Death from any cause.


Proportion of patients achieving improvement in bone marrow fibrosis of ≥1 grade from baseline Frequency, duration and severity of adverse events, abnormalities in vital signs and laboratory test values, including ECG data.


Dosing of Compound a and/or Ruxolitinib


Compound A may be administered orally at a total daily dose of 100-300 mg, for example, 100 mg, 200 mg, or 300 mg. preferably administered once daily. Preferably, Compound A is administered at a total daily dose of 100 mg or 200 mg, preferably administered once a day.


Ruxolitinib may be administered orally at a total daily dose of 10 to 50 mg. For example, ruxolitinib may be administered at a dose of 5-25 mg, administered twice daily.


Preferably, ruxolitinib is administered at a dose of 15 mg BID, or 20 mg BID (total daily dose of 30 mg or 40 mg).


As part of the combination therapy, the patient is preferably on a stable ruxolitinib dose for at least 8 weeks prior to first dose of combination therapy. The ruxolitinib dose may vary between 5 mg BID and 25 mg BID.


Thus in one embodiment, the present invention provides for the treatment of a patient suffering from an MPN, wherein the patient has been previously treated with ruxolitinib, or a pharmaceutically acceptable salt thereof.


For example, the total daily dose of ruxolitinib may be determined as follows.


For patients suffering from myelofibrosis, the starting dose of ruxolitinib may be based on patient's baseline platelet count:

    • (i) Greater than 200×109/L: 20 mg given orally twice daily;
    • (ii) 100×109/L to 200×109/L: 15 mg given orally twice daily;
    • (iii) 50×109/L to less than 100×109/L: 5 mg given orally twice daily.


Complete blood counts are monitored every 2 to 4 weeks until doses are stabilized, and then as clinically indicated. Dosing may be interrupted or adjusted for thrombocytopenia.


For patients suffering from Polycythemia Vera (2.2), the starting dose of Jakafi is 10 mg given orally twice daily. Doses of ruxolitinib may be increased in 5 mg twice daily increments to a maximum of 25 mg twice daily.

Claims
  • 1. A method of treating myeloproliferative neoplasm (MPN) in a patient, comprising administering an ERK1/2 inhibitor in combination with at least one further active agent to the patient, wherein the at least one further active agent is a JAK1/JAK2 inhibitor, a JAK2/FLT3 inhibitor, a JAK2V617F inhibitor, a JAK2 inhibitor, JAK1 inhibitor or a JAK2/Src inhibitor.
  • 2. The method of claim 1 wherein the myeloproliferative neoplasm is selected from myelofibrosis (MF), essential thrombocythemia (ET), polycythemia vera (PV) and combinations thereof.
  • 3. The method of claim 2, wherein the myelofibrosis comprises is primary myelofibrosis (PMF), post-essential thrombocythemia myelofibrosis (PET-MF) or post-polycythemia vera myelofibrosis (PPV-MF).
  • 4. The method of claim 3, wherein said patient has thrombocytopenia associated with myelofibrosis.
  • 5. The method of claim 3, wherein said patient has neutropenia associated with myelofibrosis.
  • 6. The method of claim 3, wherein said patient has a peripheral blood platelet count of less than or equal to 50,000/μL before treatment.
  • 7. The method of claim 3, wherein said patient has a peripheral blood platelet count of less than or equal to 75,000/μL before treatment.
  • 8. The method of claim 1, wherein the myeloproliferative neoplasm (MPN) is primary myelofibrosis (PMF).
  • 9. The method of claim 1 wherein median survival time increases by at least 3 months after treatment.
  • 10. The method of claim 1 wherein said patient achieves an Hb improvement of ≥2.0 g/dL or ≥1.5 g/dL after treatment.
  • 11. The method of claim 1 any one of the claims 1 to 8 wherein said patient completely responds to the treatment.
  • 12. The method of claim 1 wherein the myeloproliferative neoplasm (MPN) is newly diagnosed MF.
  • 13. (canceled)
  • 14. (canceled)
  • 15. The method of claim 1, wherein the at least one further active agent is ruxolitinib, or a pharmaceutically acceptable salt thereof.
  • 16. The method of claim 15, wherein ruxolitinib or a pharmaceutically acceptable salt thereof, is administered at a total daily dose of 10 to 50 mg, administered once or twice daily.
  • 17. The method of claim 16, wherein ruxolitinib is administered in an amount of from 5 mg twice daily to 25 mg twice daily.
  • 18. The method of claim 1, wherein the patient is receiving or has received prior therapy with ruxolitinib.
  • 19. The method of claim 18 wherein the prior therapy with ruxolitinib is administration at 5 mg twice daily, 10 mg twice daily, 15 mg twice daily, 20 mg twice daily or 25 mg twice daily.
  • 20. The method of claim 1, wherein said ERK1/2 inhibitor is 4-(3-amino-6-((1 S,3S,4S)-3-fluoro-4-hydroxycyclohexyl)pyrazin-2-yl)-N—((S)-1-(3-bromo-5-fluorophenyl)-2-(methylamino)ethyl)-2-fluorobenzamide (Compound A), or pharmaceutical acceptable salt thereof.
  • 21. The method of claim 1 wherein Compound A is administered at a total daily dose of 100-300 mg, or 200-300 mg.
  • 22. The method of claim 1 wherein Compound A is administered at a total daily dose of 100 mg or 200 mg.
  • 23. A method of treating myeloproliferative neoplasm (MPN) in a patient, comprising administering Ruxolitinib, or a pharmaceutically acceptable salt therefore in combination with 4-(3-amino-6-((1 S,3S,4S)-3-fluoro-4-hydroxycyclohexyl)pyrazin-2-yl)-N—((S)-1-(3-bromo-5-fluorophenyl)-2-(methylamino)ethyl)-2-fluorobenzamide (Compound A), or a pharmaceutically acceptable salt thereof, to the patient.
  • 24. A pharmaceutical combination of 4-(3-amino-6-((1 S,3S,4S)-3-fluoro-4-hydroxycyclohexyl)pyrazin-2-yl)-N—((S)-1-(3-bromo-5-fluorophenyl)-2-(methylamino)ethyl)-2-fluorobenzamide (Compound A), or a pharmaceutically acceptable salt thereof, and a JAK 1/2 inhibitor.
  • 25. A pharmaceutical combination according to claim 24, wherein the JAK 1/2 inhibitor is ruxolitinib, or a pharmaceutically acceptable salt thereof.
  • 26. A method of treating myeloproliferative neoplasm (MPN) in a patient, comprising administering the pharmaceutical combination according to claim 24 to the patient.
PCT Information
Filing Document Filing Date Country Kind
PCT/IB2021/059203 10/7/2021 WO
Provisional Applications (1)
Number Date Country
63089172 Oct 2020 US