NMS 122 COMBINATION OF A MPS1 INHIBITOR AND IMMUNE CHECKPOINT INHIBITORS, USES AND PHARMACEUTICAL COMPOSITIONS THEREOF Field of the invention The present invention relates to a combination of a MPS1 (Monopolar Spindle 1) kinase inhibitor and immune checkpoint inhibitors. More particularly, the invention relates to a combination of N-(2,6-diethylphenyl)-8-({4-[4- (dimethylamino)piperidin-1-yl]-2-methoxyphenyl}amino)-1-methyl-4,5-dihydro-1H-pyrazolo[4,3-h]quinazoline-3- carboxamide or one of its salts with a pharmaceutically acceptable acid or base and immune checkpoint inhibitors. The invention also relates to the use of said combination in the treatment of cancer, in particular hepatocellular carcinoma (HCC). Also provided are pharmaceutical formulations suitable for the administration of such combinations. Background of the invention Cancer is a major threat to global public health and it is still a leading cause of death worldwide. Therefore, despite recent advances in therapy, there is an urgent medical need for the development of more effective therapeutic treatments. Monopolar Spindle 1 (MPS1) kinase, also known as Tyrosine and Serine/Threonine kinase (TTK), is a core protein of the spindle assembly checkpoint (SAC), which is a signaling cascade that prevents chromosome missegregation by arresting the cell cycle in mitosis until all chromosomes are properly attached to the mitotic spindle and aligned at the metaphase plate. Thus, the SAC ensures healthy proliferation and precise division in cells. Many cancer cells have a weakened SAC response that may contribute to malignancy. However, more severe disabling of the SAC generates a level of chromosome instability that exceeds the adaptation capacity of cancer cells and is a potential anticancer strategy. Many tumors are shown to overexpress MPS1, which correlates with worse prognosis. Inhibition of MPS1 activity in cancer cells causes severe chromosome missegregation, cell death and tumor growth inhibition. N-(2,6-diethylphenyl)-8-({4-[4-(dimethylamino)piperidin-1-yl]-2-methoxyphenyl}amino)-1-methyl-4,5-dihydro-1H- pyrazolo[4,3-h]quinazoline-3-carboxamide is a compound represented by the formula (I):
that was first described in protein kinase in the low nano molar range and is thus useful in therapy as antitumor agent. Synthesis of compound of formula (I) as well as its phosphate salt is described in WO2019002454. In particular, the preferred preparation of the compound of formula (I) is that described in example 2, Step A of the International Patent Application WO2019002454. N-(2,6-diethylphenyl)-8-({4-[4-(dimethylamino)piperidin-1-yl]-2-methoxyphenyl}amino)-1-methyl-4,5-dihydro-1H- pyrazolo[4,3-h]quinazoline-3-carboxamide is presently undergoing a phase I/II clinical trial with patients having advanced solid cancers. Given the potency and selectivity of compound of formula (I) in inhibiting a critical mitotic checkpoint protein, it would be advantageous to further enhance the efficacy of this drug candidate in cancer treatment. Immuno-oncology is an innovative area of research that aims at harnessing the patient’s immune system to fight cancer. One of the most promising approaches to prevent suppression of anti-cancer immunity is the blockade of immune checkpoints, i.e. molecular pathways evolved to prevent T-cell-mediated autoimmunity, but that tumors can also exploit to their advantage. In tumors, the expression of these proteins is deregulated. For this reason, a significant line of research has focused on immune checkpoint inhibitors (ICI) to block the inhibitory receptors expressed on T cells, such as cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) and programmed cell death protein-1 (PD-1) or their corresponding ligands expressed on tumor cells, such as programmed cell death 1 ligand- 1 (PD-L1 ). (Alsaab, H. O. et at. Front. Pharmacol.8, 1-15 (2017)). Different anti-cancer drugs focused on anti CTLA-4 and PD-1/PD-L1 checkpoint inhibitors are now approved by the FDA. These include monoclonal antibodies against PD-1, PD-L1 and CTLA-4, such as atezolizumab, avelumab, cemiplimab, pembrolizumab, nivolumab, durvalumab, tremelimumab and ipilimumab. Cancer immunotherapy by blockade of immune checkpoint molecules has proved remarkable clinical efficacy across multiple cancer types, furthermore clinical trials of immune checkpoint immunotherapy have shown good results even for advanced metastatic cancers. (Alsaab, H. O. et al. Front. Pharmacol.8, 1-15 (2017)). Despite an unquestionable success, not all patients are responsive, probably because cancer can find alternative ways to escape immune surveillance. These limitations have pushed clinicians toward combinations with new antitumoral agents or new immunotherapeutic strategies that are more efficacious in inducing immune surveillance against tumors. Summary of the invention The present invention relates to a pharmaceutical combination comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, and at least one immune checkpoint inhibitor for simultaneous, sequential or separate use. Said compound of formula (I), its salts, its synthesis, its use in the treatment of cancer and pharmaceutical formulations thereof, are described in the International Patent Applications WO2009156315 and WO2019002454, the contents of which is incorporated by reference. It has now been found by the inventors of the present application that the administration of the compound of formula (I) or a pharmaceutically acceptable salt thereof in combination with an immune checkpoint inhibitor is more effective in the treatment of cancer than each single agent. Based on these results, provided herein are methods of treating cancer in a subject by administering to the subject an effective amount the compound of formula (I) or a pharmaceutically acceptable salt thereof and an effective amount of an immune checkpoint inhibitor. Also provided herein are pharmaceutical compositions comprising the compound of formula (I) or a pharmaceutically acceptable thereof and an immune checkpoint inhibitor. Brief description of the drawings Figure 1 illustrates the individual tumor growth curves of Hepa 1-6 HCC tumor-bearing mice treated with: (a) vehicle; (b) compound of formula (I) as single agent; (c) anti-PD-1 antibody as single agent; (d) combination of compound of formula (I) with anti-PD-1 antibody; (e) anti-PD-L1 antibody as single agent; (f) combination of compound of formula (I) with anti-PD-L1 antibody. Figure 2 illustrates the survival benefit of the combinations of compound of formula (I) with: (a) anti-PD-1 antibody or (b) anti-PD-L1 antibody, when compared to the vehicle and single agent treatment arms, in the Hepa 1-6 syngeneic model of HCC. Detailed description of the invention According to a first object, the present invention provides a pharmaceutical combination comprising a compound of formula (I):
or a pharmaceutically inhibitor. Pharmaceutically acceptable salts of the compound of formula (I) include the acid addition salts with inorganic or organic acids, e.g., nitric, hydrochloric, hydrobromic, sulphuric, perchloric, phosphoric, acetic, trifluoroacetic, propionic, glycolic, lactic, oxalic, malonic, malic, maleic, tartaric, citric, benzoic, cinnamic, mandelic, methanesulphonic, benzensulphonic, isethionic and salicylic acid and the like. In one preferred embodiment, the pharmaceutically acceptable salt is selected from the group consisting of phosphate, methansulphonate (tosylate) and benzensulphonate (besylate). In a more preferred embodiment, the salt is phosphate. An immune checkpoint inhibitor in accordance with the present invention may be a small-molecule organic compound or a large molecule such as a peptide or a nucleic acid. In at least one embodiment, an immune checkpoint inhibitor is an antibody or an antigen binding fragment thereof. In at least one embodiment, an immune checkpoint inhibitor is a monoclonal antibody or an antigen binding fragment thereof. As used herein, the term "antibody" means an immunoglobulin molecule that recognizes and specifically binds to a target, such as a protein, polypeptide, peptide, carbohydrate, polynucleotide, lipid, or combinations of the foregoing through at least one antigen recognition site within the variable region of the immunoglobulin molecule. As used herein, the term "antibody" encompasses intact polyclonal antibodies, intact monoclonal antibodies, antibody fragments (e.g., Fab, Fab', F(ab')2, and Fv fragments), single chain Fv (scFv) mutants, multispecific antibodies such as bispecific antibodies, chimeric antibodies, humanized antibodies, human antibodies, fusion proteins comprising an antigen. Another object of the present invention is to provide a pharmaceutical combination comprising a compound of formula (I) as defined above and at least one immune checkpoint inhibitor for use in a method of treating cancer in a subject, wherein said compound of formula (I) and the immune checkpoint inhibitor can be administered simultaneously, sequentially or separately. Another object of the present invention relates to the use of a compound of formula (I) as defined above in the preparation of a medicament for the treatment of cancer, wherein said treatment comprises simultaneously, sequentially or separately administering to a subject in need thereof a compound of formula (I) as defined above and at least one immune checkpoint inhibitors of the invention. Another object of the present invention to a method for treating cancer in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a compound of formula (I) as defined above, in combination with a therapeutically effective amount of at least one immune checkpoint inhibitor of the invention, wherein the compound of formula (I) and the immune checkpoint inhibitor can be administered the simultaneously, sequentially or separately to a subject in need thereof. Another object of the present invention relates to a combined preparation for simultaneous, separate or sequential use. The combination of the invention can be also formulated into a pharmaceutical composition prior to administration to a subject. The pharmaceutical compositions of the invention are prepared using techniques and methods known to those skilled in the art. Therefore, also object of the present invention are pharmaceutical compositions comprising a combination of a compound of formula (I) as defined above and at least one immune checkpoint inhibitor, optionally together with a pharmaceutically acceptable carrier, diluent or excipient. The term “combined preparation” as used herein defines especially a “ kit of parts” in the sense that the combination partners defined above can be dosed independently or by use of different fixed combinations with distinguished amounts of the combination partners, i.e. simultaneously or at different time points. The parts of the kit of parts can then, e.g., be administered simultaneously or chronologically staggered, that is at different time points and with equal or different time intervals for any part of the kit of parts. Very preferably, the time intervals are chosen such that the therapeutic effect of the combination of the invention on the treated disease is greater than the effect which would be obtained by use of only any one of the combination partners. The ratio of the total amounts of the combination partners to be administered in the combined preparation can be varied, e.g. in order to cope with the needs of a patient subpopulation to be treated or the needs of the single patient which different needs can be due to the particular disease, age, sex, body, weight, etc. of the patients. As used herein, a “therapeutically effective amount” of a compound refers to a sufficient amount to achieve its intended purpose. Determination of the effective amounts is well within the capability of those skilled in the art based upon the achievement of a desired effect. An effective amount will depend on factors including, but not limited to, the size of a subject and/or the degree to which the disease or unwanted condition from which a subject suffers has progressed. The effective amount will also depend on whether the compound is administered to the subject in a single dosage or periodically over time. As used herein, the terms "treatment," "treat," and "treating" refer to reversing, alleviating, ameliorating, inhibiting or slowing the progression of a cancer, reducing the likelihood of recurrence of a cancer, or one or more symptoms thereof, as described herein. As used herein, the term "subject" encompasses mammals and non-mammals. Examples of mammals include, but are not limited to, any member of the mammalian class: humans, non-human primates such as chimpanzees, and other apes and monkey species; farm animals such as cattle, horses, sheep, goats, swine; domestic animals such as rabbits, dogs and cats; laboratory animals including rodents, such as rats, mice and guinea pigs, and the like. Examples of non-mammals include, but are not limited to, birds, fish and the like. Immune checkpoint inhibitors comprise, but are limited to PD-1, PD-L1, CTLA-4, TIM3 (T cell immunoglobulin and mucin-3), OX-40 and its ligand OX40L, LAG-3 (lymphocyte activation gene-3), KIR (Killer-cell Immunoglobulin like Receptor), VISTA (V-domain Ig-containing suppressor of T cell activation), ID01 (Indoleamine 2,3- dioxygenase), TIGIT (T cell immunoglobulin and ITIM domain), BTLA (B and T lymphocyte attenuator), A2AR (Adenosine receptor A2), SIGLEC7 (Sialic acid-binding immunoglobulin-type lectin 7), GITR (Glucocorticoid- Induced TNFR family Related gene), ICOS (Inducible T-cell costimulator), NOX-2 (nicotinamide adenine dinucleotide phosphate NADPH oxidase isoform 2), Arginase I, CD276 (Cluster of Differentiation 276, also referred to as B7H4), CD27 (Cluster of Differentiation 27) and its ligand CD27 (Cluster of Differentiation 27), CD160 (Cluster of Differentiation 160) and CD39 (Cluster of Differentiation 39). In a preferred embodiment of the invention the immune checkpoint inhibitor is selected from the group consisting of PD-1 inhibitor, PD-L1 inhibitor and CTLA-4 inhibitor. In a more preferred embodiment, the PD-1 inhibitor is an anti PD-1 antibody, preferably nivolumab and pembrolizumab. In another more preferred embodiment, the PD-L1 inhibitor is and anti PD-1 antibody, preferably atezolizumab, avelumab and durvalumab. The present invention further provides a commercial package comprising, in a suitable container mean, (a) a compound of formula (I) as defined above, and (b) one or more immune checkpoint inhibitor, wherein the active ingredients are present in each case in free form or in the form of a pharmaceutically acceptable salt, together with instructions for simultaneous, separate or sequential use thereof. In a package according to the invention each of partner (a) and (b) are present within a single container mean or within distinct container means. Another embodiment of the present invention is a commercial package comprising a pharmaceutical composition or product as described above. In a preferred embodiment the cancer is selected from the group consisting of carcinomas, preferably bladder, breast, colon, kidney, liver, small cell lung cancer, esophagus, gall-bladder, ovary, pancreas, stomach, cervix, thyroid, prostate, and skin; hematopoietic tumors of lymphoid lineage, preferably acute lymphocitic leukaemia, acute lymphoblastic leukaemia, B-cell lymphoma, T-cell-lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, hairy cell lymphoma and Burkett's lymphoma; hematopoietic tumors of myeloid lineage, preferably, acute myelogenous leukemias and chronic myelogenous leukemias, myelodysplastic syndrome and promyelocytic leukaemia; tumors of mesenchymal origin, preferably fibrosarcoma and rhabdomyosarcoma; tumors of the central and peripheral nervous system, preferably astrocytoma neuroblastoma, glioma and schwannomas; melanoma; and Kaposi's sarcoma. In a more preferred embodiment, the cancer is hepatocellular carcinoma (HCC). Also included herein are pharmaceutical compositions comprising the compound of formula (I) or a pharmaceutically acceptable salt thereof and at least one immune checkpoint inhibitor as described herein optionally together with a pharmaceutically acceptable carrier, in the manufacture of a medicament for the treatment of cancer. The term "pharmaceutically acceptable carrier" refers to a non-toxic carrier, diluent, adjuvant, vehicle or excipient that does not adversely affect the pharmacological activity of the compound with which it is formulated, and which is also safe for human use. Pharmaceutically acceptable carriers that may be used in the compositions of this disclosure include, but are not limited to, ion exchangers, alumina, aluminum stearate, magnesium stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances (e.g., microcrystalline cellulose, hydroxypropyl methylcellulose, sodium carboxymethylcellulose), lactose monohydrate, sodium lauryl sulfate, polyethylene glycol, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers and wool fat. In an embodiment, an effective amount of the compound of formula (I) or a pharmaceutically acceptable salt thereof taught herein ranges from a dosage level of about 20 mg/m2/week to about 400 mg/m2/week of body surface. A dosage level of from about 20 mg/m2/week to 200 mg/m2/week constitutes a particularly suitable range. In a preferable embodiment dosage level is from 20 mg/m2/week to 150 mg/m2/week. In an embodiment, an effective amount of an immune checkpoint inhibitor taught herein ranges from about 50 to about 3000 mg per subject, alternatively from about 100 to about 1500 mg per subject. The skilled artisan will appreciate that certain factors may influence the dosage required to effectively treat a subject suffering from cancer or reduce the likelihood of recurrence of a cancer. These factors include, but are not limited to, the classification and/or severity of the disease or disorder, previous treatments, the general health and/or age of the subject and other diseases present. The compound of formula (I) or a pharmaceutically acceptable salt thereof and the immune checkpoint inhibitor, or the compositions of the present invention may be administered, for example, by oral, parenteral, sublingual, topical, rectal, nasal, buccal, vaginal, transdermal, patch, pump administration or via an implanted reservoir, and the pharmaceutical compositions would be formulated accordingly. Parenteral administration includes intravenous, intraperitoneal, subcutaneous, intramuscular, transepithelial, nasal, intrapulmonary, intrathecal, rectal and topical modes of administration. Parenteral administration can be by continuous infusion over a selected period of time. Experimental part and results Example 1. In vivo antitumor activity of compound of formula (I) Hepa 1-6 murine HCC cancer cells were cultured in DMEM (Dulbeco’s Modified Eagle Medium), supplemented with 10% heat inactivated fetal bovine serum at 37°C in an atmosphere of 5% CO2 in air. The cells in exponential growth phase were harvested and quantitated by cell counter before inoculation in C57BL/6 mice. Each mouse was inoculated subcutaneously in the right flank with 5x106 Hepa 1-6 cells in 0.1 ml of PBS for tumor development. Eight days after cell injection, animals having volume of about 95 mm3 were randomized and allocated to 6 experimental groups (n=10 mice/group) for treatment with vehicle, compound of formula (I), anti-PD-1 antibody (clone RMP1-14), anti-PD-L1 antibody (clone 10F.9G2) or the combination of compound of formula (I) with anti- PD-1 or anti-PD-L1 antibodies. compound of formula (I) was administered intravenously at 13 mg/kg every 4 days (Q4D) for 10 doses (Days 0, 4, 8, 12, 16, 20, 24, 28, 32 and 36). Anti-PD-1 antibody and anti-PD-L1 antibody were administered intraperitoneally at 200 µ gr/mouse biweekly for two weeks (Days 0, 3, 7 and 10). During the study, animals were checked daily for morbidity and mortality. Effect of treatment on tumor growth and body weight was evaluated three times per week. Mortality and observed clinical signs were recorded for individual animals. Tumors were measured in two dimensions using a caliper and the tumor volume was expressed in mm3 using the formula: V=(L x W2)/2, where V is tumor volume, L is tumor length (the longest tumor dimension) and W is tumor width (the longest tumor dimension perpendicular to L). Dosing as well as tumor and body weight were conducted in a Laminar Flow Cabinet. The tumor volumes and body weights were measured by using StudyDirectorTM software (version 3.1.399.19). Animals were followed until Day 96 and individual mice were euthanized when the ethical endpoint was reached (body weight loss over 20% relative to the weight on the first day of treatment, or tumor volume exceeding 3000 mm3). Efficacy was evaluated by survival time (death or ethical endpoint reached), and tumor free mice at the end of the study. The median survival times for the vehicle treated group and the group treated with compound of formula (I) were 40 days and 63 days, respectively. Tumor free mice were not observed in these groups (Fig.1, Fig.2, Table 1 and Table 2). Treatment with anti-PD-1 antibody was very effective in this study and resulted in tumor regressions and a strong prolongation of survival. At the end of the study, the groups treated with anti-PD-1 antibody single agent or the combination anti-PD-1 antibody with compound of formula (I) had not yet reached the median survival time. The therapeutic benefit of the combination anti-PD-1 antibody with compound of formula (I) is visible in Fig.1, which shows the tumor growth in individual mice of each treatment group. In addition, the combination group resulted in 8 tumor free mice out of 10 mice, whereas in the anti-PD-1 antibody single agent group only 6 out of 10 mice remained tumor free at the end of the study (Fig.1, Fig.2 and Table 1). Treatment with anti-PD-L1 antibody also induced tumor regressions and prolongation of survival, which was significantly improved with the combination anti-PD-L1 antibody with compound of formula (I). The median survival time for the group treated with anti-PD-L1 antibody single agent was 63 days, whereas animals in the group treated with the combination anti-PD-L1 antibody with compound formula (I), it was >94.5 days. Compared to the vehicle treated group, compound of formula (I) and anti-PD-L1 antibody single agent groups both prolonged the survival in 23 days. Interestingly, the combination of anti-PD-L1 antibody and compound of formula (I) prolonged the survival in more than 54.5 days, what indicates that the combination is synergistic. (Fig.1, Fig.2 and Table 2). Moreover, in the combination compound of formula (I) with anti-PD-L1 antibody there were 3 out of 10 mice tumor free at the end of the study, whereas in the group treated with anti-PD-L1 antibody single agent there were 2 out of 10 mice tumor free (Table 2).  Table 1 Table 2 * Log-Rank (Mantle Cox) test ( *p≤0.05, **p≤0.01, ***p≤0.001)