WO2022219128A1 - In vitro tests allowing to identify the potential for mdm2 inhibitors to induce the selection of mutations in patients suffering from a myeloproliferative neoplasm - Google Patents

In vitro tests allowing to identify the potential for mdm2 inhibitors to induce the selection of mutations in patients suffering from a myeloproliferative neoplasm Download PDF

Info

Publication number
WO2022219128A1
WO2022219128A1 PCT/EP2022/060039 EP2022060039W WO2022219128A1 WO 2022219128 A1 WO2022219128 A1 WO 2022219128A1 EP 2022060039 W EP2022060039 W EP 2022060039W WO 2022219128 A1 WO2022219128 A1 WO 2022219128A1
Authority
WO
WIPO (PCT)
Prior art keywords
mutations
mutated
mdm2
cells
mpn
Prior art date
Application number
PCT/EP2022/060039
Other languages
French (fr)
Inventor
Bruno Cassinat
Nabih MASLAH
Emmanuelle Verger
Jean-Jacques KILADJIAN
Stéphane GIRAUDIER
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
Assistance Publique-Hôpitaux De Paris (Aphp)
Université Paris Cité
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by INSERM (Institut National de la Santé et de la Recherche Médicale), Assistance Publique-Hôpitaux De Paris (Aphp), Université Paris Cité filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Publication of WO2022219128A1 publication Critical patent/WO2022219128A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/502Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects
    • G01N33/5023Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects on expression patterns
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57426Specifically defined cancers leukemia
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/22Haematology
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the present invention is in the field of medicine, in particular oncology.
  • BCR-ABL1 negative myeloproliferative neoplasms form a group of disorders characterized by the acquisition of phenotype-driving genetic lesions in hematopoietic stem cells, including mutations in th eJAK2 (JAK2 V617F and exon 12), CALR and MPL genes.
  • MPNs are heterogeneous with a chronic phase inconstantly followed by evolution to more aggressive diseases like myelofibrosis or acute myeloid leukemia (AML). This heterogeneity is likely due to the acquisition of numerous additional mutations during the course of the disease that have been reported to impact the clinical outcome (1-4).
  • TP53 TP53 protein
  • the p53 protein is considered to be a main barrier to cancer development because of its multiple anti -oncogenic functions (7-9).
  • Acquired mutations in TP53 leading to inactive forms of the protein are among the most frequent mutations in all types of cancer and are considered key events in the transformation of chronic MPN to acute leukemia.
  • MDM2 an E3 ubiquitin ligase
  • MDM2 inhibitors have been developed, the best described being nutlins, a family of small molecules blocking the p53-MDM2 interactions leading to stabilization of p53 (15). These drugs have been evaluated in patients with solid cancers and hematological malignancies with promising results. Although the impact of nutlins on wild- type p53 is well documented, the differential impact of these drugs on mutant vs wild-type proteins is not known.
  • the present invention relates to methods of determining whether a patient suffering from a myeloproliferative neoplasm is at risk of developing at least one ZP53-mutated tumoral clone under therapy with a MDM2 inhibitor.
  • the Inventors developed an in vitro assay using patient’ cells which confirmed a direct effect of this MDM2 inhibitor on clonal selection which has the potential to predict the clonal evolution in patients prior to treatment. They also used an innovative single cell DNA next generation sequencing (NGS) approach to decipher the molecular context of ZP53-mutated cells emergence.
  • NGS next generation sequencing
  • the present invention relates to a method of determining whether a patient suffering from a myeloproliferative neoplasm is at risk of developing at least one ZP53-mutated tumoral clone under therapy with a MDM2 inhibitor comprising the steps of i) culturing a sample of hematopoietic stem and progenitors cells (HSPC) obtained from the patient in presence of an amount of the MDM2 inhibitor for at least 5 days, and ii) detecting the presence of TP53- mutated tumoral clones wherein detection of said clones indicates that the patient is at risk of developing ZP53-mutated tumoral clones.
  • HSPC hematopoietic stem and progenitors cells
  • myeloproliferative neoplasm or “MPN” has its general meaning in the art and include polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF) that are a diverse but inter-related group of clonal disorders of pluripotent hematopoietic stem and progenitor cells that share a range of biological, pathological and clinical features including the relative overproduction of one or more cell types from myeloid origin with growth factor independency/hypersensitivity, marrow hypercellularity, extramedullary hematopoiesis, spleno- and hepatomegaly, and thrombotic and/or hemorrhagic diathesis.
  • PV polycythemia vera
  • ET essential thrombocythemia
  • PMF primary myelofibrosis
  • IWG-MRT myeloproliferative neoplasms research and treatment
  • the patients harbour at least one mutation in JAK2 , MPL, or CALR.
  • Mutations in the JAK2 e.g. JAK2 V617F and exon 12
  • CALR and MPL genes are characteristic of an acquisition of phenotype-driving genetic lesions in hematopoietic stem cells in BCR-ABL1 negative myeloproliferative neoplasms (MPNs) disorders.
  • JAK2 has its general meaning in the art and refers to the Janus Kinase 2 protein.
  • the amino acid sequence of human JAK2 is well known in the art. Human JAK2 sequences are, for example, represented in the NCBI database (www.ncbi.org; www.ncbi.nlm.nih.gov), for example, under accession number NP 004963.
  • Typical myeloproliferative neoplasm associated mutation is the JAK2 V617F mutation which refers to the point mutation (1849 G for T) in exon 14, which causes the substitution of phenylalanine for valine at codon 617 in the JAK homology JH2 domain.
  • JAK2 mutations include exon 12 mutations which can be substitutions, deletions, insertions and duplications, and all occur within a 44 nucleotides region in the JAK2 gene which encompasses amino acids 533-547 at the protein level.
  • MPL has its general meaning in the art and refers to the MPL proto-oncogene, thrombopoietin receptor.
  • the amino acid sequence of human MPL is well known in the art. Human MPL sequences are, for example, represented in the NCBI database (www.ncbi.org; www.ncbi.nlm.nih.gov), for example, under accession number NP_005364.1.
  • Typical MPL mutation include MPLS505N; MPLW515L, MPLW515K and the rare MPLW515A, MPLW515R and MPLW515S mutations.
  • the tryptophan residue (W) in position 515 at the intracellular juxtamembrane boundary normally inhibits dimerization of the Mpl transmembrane helix and thereby prevents receptor self-activation.
  • Replacing W515 with another amino acid, for example, leucine, lysine, or arginine leads to loss of this inhibition and results in a constitutively active Mpl.
  • Alternative mutations have also been reported in rare cases including V501A, S505C, A506T, V507I, G509C, L510P, R514K and R519T.
  • CALR has its general meaning in the art and refers to the calreticulin which is a multifunctional protein that acts as a major Ca (2+) -binding (storage) protein in the lumen of the endoplasmic reticulum.
  • the amino acid sequence of human CALR is well known in the art. Human CALR sequences are, for example, represented in the NCBI database (www.ncbi.org; www.ncbi.nlm.nih.gov), for example, under accession number NP 004334.1.
  • Typical, myeloproliferative neoplasm-associated mutations are insertions, deletions or insertions and deletions (indels) and occur in exon 9 resulting in a +1 base pair frame-shift of the coding sequence and generation of a translated protein with a novel C- terminus that lacks a retrieval sequence (KDEL) typical of normal CALR and other endoplasmic reticulum resident proteins.
  • KDEL retrieval sequence
  • Specific examples include CALRdel52/type I; c.l092_1143del; L367 fs*46 or a 5-bp insertion CALRins5/type II; cl 154 1155insTTGTC; K385 fs*47.
  • P53 has its general meaning in the art and refers the tumor human suppressor protein containing transcriptional activation, DNA binding, and oligomerization domains.
  • the P53 protein responds to diverse cellular stresses to regulate expression of target genes, thereby inducing cell cycle arrest, apoptosis, senescence, DNA repair, or changes in metabolism. Mutations in this gene are associated with a variety of human cancers, including hereditary cancers such as Li-Fraumeni syndrome, since P53 is an anti oncogene protein which has an essential role in the protection of cells against cancer.
  • LP5J refers to the gene encoding P53 protein.
  • NCBI reference gene ID is 7151 and is accessible under Ensembl:ENSG00000141510 MIM:191170.
  • TP53 mutation has its general meaning in the art and refers to any mutation which results into a dysfunction of the protein leading to the loss of its transcriptional activity associated with a negative effect on the wild type protein in heterozygous status.
  • P53 loss of function mutations have fully been exemplified in the prior art and thus the skilled man in the art can easily identify TP53 mutations (Petit jeans A, Mathe E, Kato S, Ishioka C, Tavtigian SV, Hainaut P, Olivier M. Impact of mutant P53 functional properties on TP53 mutation patterns and tumor phenotype: lessons from recent developments in the IARC TP53 database.Hum Mutat. 2007 Jun;28(6):622-9) (http://P53.iarc.fr/).
  • TP53 mutations are mainly missense mutations. Examples of TP53 mutations include but are not limited to R175H, Y163C, C242Y, Y243H, G245S, M246V, R248W, R248Q, R273H, C277Y and C277F. TP 53 mutations are major oncogenic events, strongly associated with the development of many cancers.
  • tumoral clone has its general meaning in the art and refers to a population of tumor cells formed by repeated division from a common cell.
  • TP53 mutated tumoral clone refers to a tumoral clone that harbors at least one TP 53 mutation.
  • MDM2 has its general meaning in the art and refers to E3 ubiquitin-protein ligase Mdm2 P53 binding protein that is a P53-associated protein (Oliner, J. D., et al., Nature 358 (1992) 80-83; Momand, J., et al., Cell 69 (1992) 1237-1245; Chen, J., et al., Mol. Cell. Biol. 13 (1993) 4107-4114; and Bueso-Ramos C. E., et al., Blood 82 (1993) 2617-2623). It is a nuclear phosphoprotein that binds and inhibits transactivation by tumor protein P53, as part of an autoregulatory negative feedback loop. MDM2 also regulates the wild-type P53 protein function by inducing its degradation.
  • MDM2 inhibitor has its general meaning in the art and refers to therapeutic agents that inhibit the MDM2-P53 interaction. MDM2 inhibitors are usually used as drugs in the field of cancer to stabilize wild-type P53 protein and promote an anti -oncogenic activity.
  • a benzodiazepinedione a sulphonamide, a chromenotriazolopyrimidine, a morpholinone, a piperidinone, a terphenyl, a chalcone, a pyrazole, an imidazole, an imidazole-indole, an isoindolinone, a pyrrolidinone, a piperidine, a naturally derived prenylated xanthone, a stapled peptide, a benzothi azole, and stictic acid.
  • MDM2 inhibitors examples include chalcone (Stoll et al, (2001) Biochemistry 40: 336-44) and sulfonamide (Galatin et al., (2004) J. Med. Chem. 47: 4163-4165).
  • MDM2 inhibitors examples include chalcone (Stoll et al, (2001) Biochemistry 40: 336-44) and sulfonamide (Galatin et al., (2004) J. Med. Chem. 47: 4163-4165).
  • MDM2 inhibitors examples include a et al., (2014) Nature Revs. 13: 217-236, incorporated herein by reference in its entirety.
  • Methods for identifying MDM2 inhibitors are well known in the art (e.g. Han AR, Durgannavar T, Ahn D, Chung SJ. A FRET-Based Fluorescent Probe to Screen Anticancer Drugs, Inhibiting p73 Binding to MDM2. Chembiochem.
  • a screening assay using a biosensor involving the physical principle of surface plasm on resonance (SPR) may be performed.
  • P53 may be immobilized on a surface and MDM2 may be put in contact concomitantly with the potential inhibitor.
  • the potential inhibitor is a MDM2 inhibitor, the percentage of binding between p53 and MDM2 decrease in dose-dependent manner.
  • a crystal structure can also be determined.
  • the MDM2 inhibitor is a nutlin.
  • the term “nutlin” refers to cis-imidazoline analogs that inhibit the interaction between MDM2 and tumor suppressor P53, and which were discovered by screening a chemical library by Vassilev et al., (2004) Science 303: 844-848. Nutlin- 1, Nutlin-2 and Nutlin-3 were all identified in the same screen.
  • Nutlin-3 ((( ⁇ )-4-[4,5-Bis(4-chlorophenyl)-2-(2-isopropoxy-4-methoxy- phenyl)-4,5-dihydro-imidazole-l-carbonyl]-piperazin-2-one) is the compound most commonly used in anti-cancer studies. Inhibiting the interaction between MDM2 and P53 stabilizes P53 and is thought to selectively induce a growth-inhibiting state called senescence in cancer cells.
  • the term “nutlin” as used herein further refers to enantiomers and stereoisomers.
  • nutlin may further refer to “second-generation” nutlin” derivatives such as, but not limited to, RG7388 (ChemieTek, Indianapolis, IN) (described by Ding et al. (2013) JMed Chem. 56: 5979-5983 and incorporated herein by reference in its entirety) and to derivatives described in, for example, US Patent Applications 20150211073 and 20170008904.
  • second-generation nutlin derivatives such as, but not limited to, RG7388 (ChemieTek, Indianapolis, IN) (described by Ding et al. (2013) JMed Chem. 56: 5979-5983 and incorporated herein by reference in its entirety) and to derivatives described in, for example, US Patent Applications 20150211073 and 20170008904.
  • the MDM2 inhibitor is Idasanutlin.
  • Idasanutlin has its general meaning in the art and refers to 4-[(2R,3S,4R,5S)-3-(3-chloro-2- fluorophenyl)-4-(4-chloro-2-fluorophenyl)-4-cyano-5-(2,2-dimethylpropyl)pyrrolidine-2- amido]-3-methoxybenzoic acid.
  • treatment refers to both prophylactic or preventive treatment as well as curative or disease modifying treatment, including treatment of subjects at risk of contracting the disease or suspected to have contracted the disease as well as subjects who are ill or have been diagnosed as suffering from a disease or medical condition, and includes suppression of clinical relapse.
  • the treatment may be administered to a subject having a medical disorder or who ultimately may acquire the disorder, in order to prevent, cure, delay the onset of, reduce the severity of, or ameliorate one or more symptoms of a disorder or recurring disorder, or in order to prolong the survival of a subject beyond that expected in the absence of such treatment.
  • therapeutic regimen is meant the pattern of treatment of an illness, e.g., the pattern of dosing used during therapy.
  • a therapeutic regimen may include an induction regimen and a maintenance regimen.
  • the phrase “induction regimen” or “induction period” refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the initial treatment of a disease.
  • the general goal of an induction regimen is to provide a high level of drug to a subject during the initial period of a treatment regimen.
  • An induction regimen may employ (in part or in whole) a "loading regimen", which may include administering a greater dose of the drug than a physician would employ during a maintenance regimen, administering a drug more frequently than a physician would administer the drug during a maintenance regimen, or both.
  • maintenance regimen refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the maintenance of a subject during treatment of an illness, e.g., to keep the subject in remission for long periods of time (months or years).
  • a maintenance regimen may employ continuous therapy (e.g., administering a drug at a regular intervals, e.g., weekly, monthly, yearly, etc.) or intermittent therapy (e.g., interrupted treatment, intermittent treatment, treatment at relapse, or treatment upon achievement of a particular predetermined criteria [e.g., disease manifestation, etc.]).
  • risk in the context of the present invention, relates to the probability that an event will occur over a specific time period and can mean a subject's "absolute” risk or “relative” risk.
  • Absolute risk can be measured with reference to either actual observation post-measurement for the relevant time cohort, or with reference to index values developed from statistically valid historical cohorts that have been followed for the relevant time period.
  • Relative risk refers to the ratio of absolute risks of a subject compared either to the absolute risks of low risk cohorts or an average population risk, which can vary by how clinical risk factors are assessed.
  • Odds ratios the proportion of positive events to negative events for a given test result, are also commonly used (odds are according to the formula p/(l-p) where p is the probability of event and (1- p) is the probability of no event) to no- conversion.
  • "Risk evaluation,” or “evaluation of risk” in the context of the present invention encompasses making a prediction of the probability, odds, or likelihood that an event or disease state may occur, the rate of occurrence of the event or conversion from one disease state to another. Risk evaluation can also comprise prediction of future clinical parameters, traditional laboratory risk factor values, or other indices of relapse, either in absolute or relative terms in reference to a previously measured population.
  • the methods of the present invention may be used to make continuous or categorical measurements of the risk of conversion, thus diagnosing and defining the risk spectrum of a category of subjects defined as being at risk of conversion.
  • the invention can be used to discriminate between normal and other subject cohorts at higher risk.
  • the method of the present invention is thus particularly suitable for determining whether the patient is at risk of developing a secondary acute leukemia.
  • acute leukemia means a disease that is characterized by a rapid increase in the numbers of immature blood cells that transform into malignant cells, rapid progression and accumulation of the malignant cells, which spill into the bloodstream and spread to other organs of the body.
  • the method of the present invention is particularly suitable for determining whether a patient will be eligible for the therapy with the MDM2 inhibitor.
  • the method of the present invention is particularly suitable also for the physician to decide that the patient shall be carefully monitored during and after the treatment so as to select the most accurate clinical decisions.
  • hematopoietic stem and progenitor cells or “HSPCs” refers to blood cells that have the capacity to self-renew and to differentiate into precursors of blood cells. These precursor cells are immature blood cells that cannot self-renew and must differentiate into mature blood cells.
  • HSPCs are typically CD34 + but display a number of phenotypes, such as Lin-CD34 + CD38XD90 + CD45RA , Lin-CD34 + CD38XD90XD45RA Lin-CD34 + CD38 + IL-3aloCD45RA , and Lin-CD34 + CD38 + CD10 + (Daley et al., Focus 18:62- 67, 1996; Pimentel, E., Ed., Handbook of Growth Factors Vol. IP: Hematopoietic Growth Factors and Cytokines, pp. 1-2, CRC Press, Boca Raton, Fla., 1994).
  • the hematopoietic stem and progenitor cells are isolated form peripheral blood cells.
  • Any culture medium suitable for growth, survival and differentiation of hematopoietic stem and progenitor cells may be used.
  • it consists of a base medium containing nutrients (a source of carbon, amino acids), a pH buffer and salts, which can be supplemented with serum of human or other origin and/or growth factors and/or antibiotics to which cytokines are added.
  • the base medium can be RPMI 1640, DMEM, IMDM, X-VIVO or AIM- V medium, all of which are commercially available standard media.
  • the culture medium comprises an amount of IL-3 (i.e. interleukin-3), an amount of SCF (i.e. stem cell factor), an amount of Flt-3 ligand (i.e. FMS- like tyrosine kinase 3), an amount of IL-6 (i.e. interleukin-6) and an amount of TPO (i.e. thrombopoietin).
  • IL-3 i.e. interleukin-3
  • SCF i.e. stem cell factor
  • Flt-3 ligand i.e. FMS- like tyrosine kinase 3
  • an amount of IL-6 i.e. interleukin-6
  • TPO i.e. thrombopoietin
  • the present invention relates to a method of determining whether a patient suffering from a myeloproliferative neoplasm (MPN) is at risk of developing at least one ZP53-mutated tumoral clone under therapy with a MDM2 inhibitor comprising the steps of i) culturing in a culture medium comprising an amount of IL-3 (i.e. interleukin-3), an amount of SCF (i.e. stem cell factor), an amount of Flt-3 ligand (i.e. FMS-like tyrosine kinase 3), an amount of IL-6 (i.e. interleukin-6) and an amount of TPO (i.e.
  • IL-3 i.e. interleukin-3
  • SCF i.e. stem cell factor
  • Flt-3 ligand i.e. FMS-like tyrosine kinase 3
  • IL-6 i.e. interleukin-6
  • TPO i.e.
  • thrombopoietin a sample of hematopoietic stem and progenitors cells (HSPC) obtained from the patient in presence of an amount of the MDM2 inhibitor for at least 10 days, and ii) detecting the presence and quantifying of ZP53-mutated tumoral clones wherein detection of said clones indicates that the patient is at risk of developing ZP53-mutated tumoral clones and the quantification in comparison with non-treated condition allows to predict the expansion of a pre-existing TP53- mutated clone.
  • HSPC hematopoietic stem and progenitors cells
  • IL-3 is used in an amount comprised between 40ng/mL and 60ng/mL and is preferably about 50 ng/mL.
  • SCF is used in an amount comprised between 80ng/mL and 120ng/mL and is preferably about 100 ng/mL.
  • IL-6 is used in an amount comprised between 40ng/mL and 60ng/mL and is preferably about 50 ng/mL.
  • Flt-3 ligand is used in an amount comprised between 80ng/mL and 120ng/mL and is preferably about 100 ng/mL.
  • TPO is used in an amount comprised between lOng/mL and 30ng/mL and is preferably about 20 ng/mL.
  • the MDM2 inhibitor is used in an amount comprised between lOnM and 30nM and is preferably about 20nM.
  • the culture of the hematopoietic stem and progenitor cells is carried out for about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 days. In some embodiments, the culture of the hematopoietic stem and progenitor cells is carried out for at least 10 days.
  • detection of the TP53 mutated tumoral clones is performed by any method well known in the art. In some embodiments, the detection of the TP53 mutated tumoral clones is performed by sequencing. Typically, nucleic acids are extracted and purified from the hematopoietic stem and progenitor cells at the end of the culture and sequencing is performed. According to the present invention, a plurality of reads is thus obtained.
  • the term “read” refers to a sequence read from a portion of a nucleic acid sample. Typically, a read represents a short sequence of contiguous base pairs in the sample.
  • the read may be represented symbolically by the base pair sequence in A, T, C, and G of the sample portion, together with a probabilistic estimate of the correctness of the base (quality score).
  • the term “sequencing” generally means a process for determining the order of nucleotides in a nucleic acid. A variety of methods for sequencing nucleic acids is well known in the art and can be used. In some embodiments, next generation sequencing is carried out. As used herein, the term “next generation sequencing” has its general meaning in the art and refers to sequencing technologies having increased throughput as compared to traditional Sanger- and capillary electrophoresis-based approaches, for example with the ability to generate hundreds of thousands or millions of relatively short sequence reads at a time.
  • Next-generation sequencers are well known in the art and can include a number of different sequencers based on different technologies, such as Illumina (Solexa) sequencing, Roche 454 sequencing, Ion torrent sequencing, SOLiD sequencing, and the like.
  • Illumina Solexa
  • Roche 454 sequencing Ion torrent sequencing
  • SOLiD sequencing SOLiD sequencing
  • An example of a sequencing technology that can be used in the present methods is the Illumina platform.
  • the Illumina platform is based on amplification of DNA on a solid surface (e.g., flow cell) using fold-back PCR and anchored primers (e.g., capture oligonucleotides). For sequencing with the Illumina platform, DNA is thus fragmented, and adapters are added to both terminal ends of the fragments (see the preceding step).
  • DNA fragments are attached to the surface of flow cell channels by capturing oligonucleotides which are capable of hybridizing to the adapter ends of the fragments.
  • the DNA fragments are then extended and bridge amplified. After multiple cycles of solid-phase amplification followed by denaturation, an array of millions of spatially immobilized nucleic acid clusters or colonies of single-stranded nucleic acids are generated. Each cluster may include approximately hundreds to a thousand copies of single-stranded DNA molecules of the same template.
  • the Illumina platform uses a sequencing-by-synthesis method where sequencing nucleotides comprising detectable labels (e.g., fluorophores) are added successively to a free 3 'hydroxyl group.
  • a laser light of a wavelength specific for the labeled nucleotides can be used to excite the labels.
  • An image is captured and the identity of the nucleotide base is recorded. These steps can be repeated to sequence the rest of the bases. Sequencing according to this technology is described in, for example, U.S. Patent Publication Application Nos. 2011/0009278, 2007/0014362, 2006/0024681, 2006/0292611, and U.S. Pat. Nos. 7,960,120, 7,835,871, 7,232,656, and 7,115,200, each of which is incorporated herein by reference in its entirety.
  • the method of the present invention further comprises the steps of i) implementing an algorithm on data that results from the sequencing as to obtain an algorithm output; and ii) determining the probability that the patient will develop at least one TP53 mutated tumoral clone under therapy with the MDM2 inhibitor.
  • algorithm is any mathematical equation, algorithmic, analytical or programmed process, or statistical technique that takes one or more continuous parameters and calculates an output value, sometimes referred to as an “index” or “index value.”
  • algorithms include sums, ratios, and regression operators, such as coefficients or exponents, biomarker value transformations and normalizations (including, without limitation, those normalization schemes based on clinical parameters, such as gender, age, or ethnicity), rules and guidelines, statistical classification models, and neural networks trained on historical populations.
  • pattern recognition features including established techniques such as cross-correlation, Principal Components Analysis (PCA), factor rotation, Logistic Regression (LogReg), Linear Discriminant Analysis (LDA), Eigengene Linear Discriminant Analysis (ELDA), Support Vector Machines (SVM), Random Forest (RF), Recursive Partitioning Tree (RPART), as well as other related decision tree classification techniques, Shrunken Centroids (SC), StepAIC, Kth-Nearest Neighbor, Boosting, Decision Trees, Neural Networks, Bayesian Networks, Support Vector Machines, and Hidden Markov Models, among others.
  • Other techniques may be used in survival and time to event hazard analysis, including Cox, Weibull, Kaplan-Meier and Greenwood models well known to those of skill in the art.
  • the method of the present invention comprises the use of a machine learning algorithm.
  • the machine learning algorithm may comprise a supervised learning algorithm.
  • supervised learning algorithms may include Average One- Dependence Estimators (AODE), Artificial neural network (e.g., Backpropagation), Bayesian statistics (e.g., Naive Bayes classifier, Bayesian network, Bayesian knowledge base), Case- based reasoning, Decision trees, Inductive logic programming, Gaussian process regression, Group method of data handling (GMDH), Learning Automata, Learning Vector Quantization, Minimum message length (decision trees, decision graphs, etc.), Lazy learning, Instance-based learning Nearest Neighbor Algorithm, Analogical modeling, Probably approximately correct learning (PAC) learning, Ripple down rules, a knowledge acquisition methodology, Symbolic machine learning algorithms, Subsymbolic machine learning algorithms, Support vector machines, Random Forests, Ensembles of classifiers, Bootstrap aggregating (bagging), and Boosting.
  • AODE Average One- Dependence Estimators
  • Supervised learning may comprise ordinal classification such as regression analysis and Information fuzzy networks (IFN).
  • supervised learning methods may comprise statistical classification, such as AODE, Linear classifiers (e.g., Fisher's linear discriminant, Logistic regression, Naive Bayes classifier, Perceptron, and Support vector machine), quadratic classifiers, k-nearest neighbor, Boosting, Decision trees (e.g., C4.5, Random forests), Bayesian networks, and Hidden Markov models.
  • the machine learning algorithms may also comprise an unsupervised learning algorithm. Examples of unsupervised learning algorithms may include artificial neural network, Data clustering, Expectation- maximization algorithm, Self-organizing map, Radial basis function network, Vector Quantization, Generative topographic map, Information bottleneck method, and IBSEAD.
  • Unsupervised learning may also comprise association rule learning algorithms such as Apriori algorithm, Eclat algorithm and FP-growth algorithm.
  • Hierarchical clustering such as Single linkage clustering and Conceptual clustering, may also be used.
  • unsupervised learning may comprise partitional clustering such as K-means algorithm and Fuzzy clustering.
  • the machine learning algorithms comprise a reinforcement learning algorithm Examples of reinforcement learning algorithms include, but are not limited to, temporal difference learning, Q-learning and Learning Automata.
  • the machine learning algorithm may comprise Data Pre-processing.
  • the output obtained by the algorithm is a score.
  • score refers to a piece of information, usually a number that conveys the result of the subject on a test.
  • a risk scoring system separates a patient population into different risk groups; herein the process of risk stratification classifies the patients into very high-risk, high- risk, intermediate-risk and low-risk groups.
  • kits or devices of the present invention comprise at least one sample collection container for sample collection. Collection devices and container include but are not limited to syringes, lancets, collection tubes.
  • the container contains the predetermined amount of the different cytokines (e.g. IL-3, SCF, IL-6, FLT-3 Ligand and TPO) and the MDM2 inhibitor.
  • the kits or devices described herein further comprise instructions for using the kit or device and interpretation of results.
  • the kit or device of the present invention further comprises a microprocessor to implement an algorithm so as to determine the probability that the patient will develop TP53 mutated tumoral clones. In some embodiments, the kit or device of the present invention further comprises a visual display and/or audible signal that indicates the probability determined by the microprocessor.
  • FIGURES are a diagrammatic representation of FIGURES.
  • Figure 1 Increase of mutant TP53 VAFs in HSPC of 7P5.?-mutated MPN patients upon MDM2 inhibitor treatment.
  • A Comparison of various TP53 mutations VAFs in CD34+ cells of MPN1 patient after 10 days with or without 20 nM of idasanutlin.
  • B Comparison of all TP 53 mutations VAFs in CD34+ cells of 8 different MPN patients after 10 days in presence or absence of idasanutlin 20 nM.
  • C Mean +/- SD of VAFs for all TP 53 mutants found in 8 MPN patients after 10 days in presence or absence of idasanutlin 20 nM or Ruxolitinib 70nM in 4 other MPN patients.
  • MPN patients were selected on the basis of having one driver mutation and at least one TP53 mutation detectable by NGS. Most of these patients also harbored mutations in other genes. Clinical and molecular characteristics are given in Table 1.
  • CD34 + cells from MPN patients after NGS analysis were prospectively cryopreserved.
  • Peripheral mononuclear cells were isolated from whole blood using Ficoll (Eurobio) and CD34 + cells sorted using a column-free immunomagnetic approach (EasySep®, StemCell).
  • 10 5 CD34 + cells were cultured in CTS StemProTM HSC expansion medium (Thermo Fisher) in which the following cytokines were added: IL-3 (50 ng/mL), SCF (100 ng/mL), TPO (20 ng/mL), IL-6 (50 ng/mL), FLT-3 Ligand (100 ng/mL).
  • Idasanutlin (provided by Roche) was added at 20 nM. After 10 days of culture, cells were harvested, DNA extracted and submitted to NGS analysis.
  • NGS analysis was used a capture-based custom NGS panel (Sophia Genetics) targeting 36 myeloid genes (ABU; ASXL1; BRAF; CALR; CBL; CCND2; CEBPA; CSF3R; CUX1; DNMT3A; ETNK1; ETV6; EZH2; FLT3; HRAS; IDH1; IDH2; IKZF1; JAK2; KIT; KRAS; MPL; NFE2; NPM1; NRAS; PTPN11; RUNX1; SETBP1; SF3B1; SH2B3; SRSF2; TET2; TP53; U2AF1; WT1; ZRSR2).
  • ABU capture-based custom NGS panel
  • CD34 + were resuspended in Tapestri® cell buffer (Mission Bio) and quantified using an automatic cell counter (Biorad).
  • Viable single cells 3,000-4,000 cells/m ⁇
  • Viable single cells 3,000-4,000 cells/m ⁇
  • PCR products were removed from individual droplets, purified with Ampure XP beads (Beckman Coulter), and used as a template for PCR to incorporate Illumina i5/i7 indices. PCR products were purified a second time, quantified via an Agilent Bioanalyzer and pooled to be sequenced. Library pools were sequenced on aNextSeq instrument (Illumina).
  • the Tapestri Insights® pipeline was used to filter variants (data not shown) and samples were included if they harbored three or more protein-encoding, non-synonymous/insertion/deletion variants and more than 1000 cells with definitive genotype for all protein-coding variants within the sample.
  • the estimated median allele dropout (ADO) rate was 8.74% (IQR: 6.5% - 10.6%, data not shown).
  • VAF allelic frequencies
  • TP53 mutations may be acquired in three distinct scenarios: either in a clone mutated for a driver gene and an epigenetic gene (TET2, DNMT3A , ASXL1 or EZH2), which is the most frequent situation (#MPN1, 2, 6), or in a clone mutated for a driver gene and a splicing factor (U2AF1 in #MPN3) or in a clone with a driver mutation only that has undergone homologous recombination (#MPN7) or not (#MPN4, 5, 8).
  • TP53 mutations may be acquired in a large array of molecular landscapes without evidence for a specific association with a particular pathway alteration, with the notable exception of the presence of driver mutations.
  • idasanutlin treatment in vitro we observed a significant increase in the VAFs of every ZP53-mutated clone.
  • MDM2 inhibitor directly favors the expansion of cells harboring TP53 mutations, an effect not observed in this study with JAK inhibition using ruxolitinib, and raises the question of the mechanism by which the inhibition of MDM2 acts on clonal selection.
  • idasanutlin treatment activates the p53 pathway and cell death in wild-type p53 cells, which would not occur in p53 mutant cells with loss of wild-type functions.
  • mutations confer gain of function mutations to p53 leading to positive selection of mutated cells.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Physics & Mathematics (AREA)
  • Medicinal Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Physics & Mathematics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (AREA)
  • Genetics & Genomics (AREA)
  • Toxicology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biophysics (AREA)
  • General Engineering & Computer Science (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

TP53 mutations are the most frequent mutations in all cancer subtypes and their acquisition are key events leading to the transformation of chronic myeloproliferative neoplasms (MPN) to leukemia. The MDM2 ubiquitin ligase is responsible for proteasome-dependent p53 degradation. MDM2 inhibitors (nutlins) may rescue p53 from degradation and have been evaluated in a variety of cancers. The inventors report herein evidence of a direct effect of MDM2 inhibition on the selection of MPN patients' cells harboring TP53 mutations. To decipher whether these mutations can arise in a specific molecular context they used a DNA single cell approach to determine the clonal architecture of TP53 mutations. They observed that: (i) clonal evolution in MPN frequently consists of sequential branching instead of linear consecutive acquisition of mutations in the same clone; (ii) TP53 mutations are late events mainly occurring in the driver clone; (iii) additional mutations don't appear to influence the selection by MDM2 inhibitor treatment. This is the first demonstration that a treatment can directly favor the emergence of potentially aggressive MPN subclones. As the molecular environment doesn't affect the potential selection of TP53 mutated cells by MDM2 inhibitors, such events could occur irrespective of the subtype of cancer.

Description

IN VITRO TESTS ALLOWING TO IDENTIFY THE POTENTIAL FOR MDM2 INHIBITORS TO INDUCE THE SELECTION OF MUTATIONS IN PATIENTS SUFFERING FROM A MYELOPROLIFERATIVE NEOPLASM
FIELD OF THE INVENTION:
The present invention is in the field of medicine, in particular oncology.
BACKGROUND OF THE INVENTION:
BCR-ABL1 negative myeloproliferative neoplasms (MPNs) form a group of disorders characterized by the acquisition of phenotype-driving genetic lesions in hematopoietic stem cells, including mutations in th eJAK2 (JAK2V617F and exon 12), CALR and MPL genes. MPNs are heterogeneous with a chronic phase inconstantly followed by evolution to more aggressive diseases like myelofibrosis or acute myeloid leukemia (AML). This heterogeneity is likely due to the acquisition of numerous additional mutations during the course of the disease that have been reported to impact the clinical outcome (1-4). Among these, the acquisition of TP53 mutations is identified in 16% of cases of MPN (5) and is associated with a poorer prognosis (6). The p53 protein is considered to be a main barrier to cancer development because of its multiple anti -oncogenic functions (7-9). Acquired mutations in TP53 leading to inactive forms of the protein are among the most frequent mutations in all types of cancer and are considered key events in the transformation of chronic MPN to acute leukemia.
Recently, anti-cancer therapies were developed to either restore the mutant p53 protein conformation (10, 11) or reinforce the wild-type p53 functions by inhibiting its degradation using MDM2 inhibitors. MDM2, an E3 ubiquitin ligase, targets p53 for degradation by the proteasome (12-14). Several MDM2 inhibitors have been developed, the best described being nutlins, a family of small molecules blocking the p53-MDM2 interactions leading to stabilization of p53 (15). These drugs have been evaluated in patients with solid cancers and hematological malignancies with promising results. Although the impact of nutlins on wild- type p53 is well documented, the differential impact of these drugs on mutant vs wild-type proteins is not known. This question is highly relevant in myeloid disorders such as MPNs where ZP53-mutated cells can emerge secondarily during the course of the disease. Based on the selective activity of nutlins on JAK2V617F mutated PV progenitor cells (16) a phase 1 trial was conducted using idasanutlin in PV patients (NCT02407080) (17). Importantly, the Inventors recently showed the expansion of 7/J53-mutated clones in some of the patients included in this trial (18). In five patients, variant allelic frequencies (VAFs) of pre-existing ZP53-mutant sub-clones increased under treatment and subsequently decreased after therapy discontinuation suggesting a drug-dependent selection of clones with leukemic potential in MPNs. Several questions arose from this observation: Does MDM2 inhibition exert a direct selective pressure allowing the emergence of ZP53-mutated clones? Does the molecular context (additional mutations) of ZP53-mutated cells impact on the selection potential of these drugs? Is it possible to better predict the risk for an individual patient to develop ZP53-mutated clones receiving idasanutlin therapy?
SUMMARY OF THE INVENTION:
As defined by the claims, the present invention relates to methods of determining whether a patient suffering from a myeloproliferative neoplasm is at risk of developing at least one ZP53-mutated tumoral clone under therapy with a MDM2 inhibitor.
DETAILED DESCRIPTION OF THE INVENTION:
In this study, the Inventors developed an in vitro assay using patient’ cells which confirmed a direct effect of this MDM2 inhibitor on clonal selection which has the potential to predict the clonal evolution in patients prior to treatment. They also used an innovative single cell DNA next generation sequencing (NGS) approach to decipher the molecular context of ZP53-mutated cells emergence.
The present invention relates to a method of determining whether a patient suffering from a myeloproliferative neoplasm is at risk of developing at least one ZP53-mutated tumoral clone under therapy with a MDM2 inhibitor comprising the steps of i) culturing a sample of hematopoietic stem and progenitors cells (HSPC) obtained from the patient in presence of an amount of the MDM2 inhibitor for at least 5 days, and ii) detecting the presence of TP53- mutated tumoral clones wherein detection of said clones indicates that the patient is at risk of developing ZP53-mutated tumoral clones.
As used herein, the term “myeloproliferative neoplasm” or “MPN” has its general meaning in the art and include polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF) that are a diverse but inter-related group of clonal disorders of pluripotent hematopoietic stem and progenitor cells that share a range of biological, pathological and clinical features including the relative overproduction of one or more cell types from myeloid origin with growth factor independency/hypersensitivity, marrow hypercellularity, extramedullary hematopoiesis, spleno- and hepatomegaly, and thrombotic and/or hemorrhagic diathesis. An international working group for myeloproliferative neoplasms research and treatment (IWG-MRT) has been established to delineate and define these conditions (see for instance Vannucchi et al, CA Cancer J. Clin., 2009, 59:171-191), and those disease definitions are to be applied for purposes of this specification. MPN has traditionally been considered to be synonymous with ‘preleukemia’ because of the increased risk of transformation into acute myelogenous leukemia (AML).
In some embodiments, the patients harbour at least one mutation in JAK2 , MPL, or CALR. Mutations in the JAK2 (e.g. JAK2V617F and exon 12), CALR and MPL genes are characteristic of an acquisition of phenotype-driving genetic lesions in hematopoietic stem cells in BCR-ABL1 negative myeloproliferative neoplasms (MPNs) disorders.
As used herein the term “JAK2” has its general meaning in the art and refers to the Janus Kinase 2 protein. The amino acid sequence of human JAK2 is well known in the art. Human JAK2 sequences are, for example, represented in the NCBI database (www.ncbi.org; www.ncbi.nlm.nih.gov), for example, under accession number NP 004963. Typical myeloproliferative neoplasm associated mutation is the JAK2V617F mutation which refers to the point mutation (1849 G for T) in exon 14, which causes the substitution of phenylalanine for valine at codon 617 in the JAK homology JH2 domain. Other examples of JAK2 mutations include exon 12 mutations which can be substitutions, deletions, insertions and duplications, and all occur within a 44 nucleotides region in the JAK2 gene which encompasses amino acids 533-547 at the protein level.
As used herein, the term “MPL” has its general meaning in the art and refers to the MPL proto-oncogene, thrombopoietin receptor. The amino acid sequence of human MPL is well known in the art. Human MPL sequences are, for example, represented in the NCBI database (www.ncbi.org; www.ncbi.nlm.nih.gov), for example, under accession number NP_005364.1. Typical MPL mutation include MPLS505N; MPLW515L, MPLW515K and the rare MPLW515A, MPLW515R and MPLW515S mutations. The tryptophan residue (W) in position 515 at the intracellular juxtamembrane boundary normally inhibits dimerization of the Mpl transmembrane helix and thereby prevents receptor self-activation. Replacing W515 with another amino acid, for example, leucine, lysine, or arginine, leads to loss of this inhibition and results in a constitutively active Mpl. Alternative mutations have also been reported in rare cases including V501A, S505C, A506T, V507I, G509C, L510P, R514K and R519T.
As used herein, the term “CALR” has its general meaning in the art and refers to the calreticulin which is a multifunctional protein that acts as a major Ca(2+) -binding (storage) protein in the lumen of the endoplasmic reticulum. The amino acid sequence of human CALR is well known in the art. Human CALR sequences are, for example, represented in the NCBI database (www.ncbi.org; www.ncbi.nlm.nih.gov), for example, under accession number NP 004334.1. Typical, myeloproliferative neoplasm-associated mutations are insertions, deletions or insertions and deletions (indels) and occur in exon 9 resulting in a +1 base pair frame-shift of the coding sequence and generation of a translated protein with a novel C- terminus that lacks a retrieval sequence (KDEL) typical of normal CALR and other endoplasmic reticulum resident proteins. Specific examples include CALRdel52/type I; c.l092_1143del; L367 fs*46 or a 5-bp insertion CALRins5/type II; cl 154 1155insTTGTC; K385 fs*47.
As used herein, the term “P53” has its general meaning in the art and refers the tumor human suppressor protein containing transcriptional activation, DNA binding, and oligomerization domains. The P53 protein responds to diverse cellular stresses to regulate expression of target genes, thereby inducing cell cycle arrest, apoptosis, senescence, DNA repair, or changes in metabolism. Mutations in this gene are associated with a variety of human cancers, including hereditary cancers such as Li-Fraumeni syndrome, since P53 is an anti oncogene protein which has an essential role in the protection of cells against cancer.
As used herein, the term “LP5J” refers to the gene encoding P53 protein. The NCBI reference gene ID is 7151 and is accessible under Ensembl:ENSG00000141510 MIM:191170.
As used the term “TP53 mutation” has its general meaning in the art and refers to any mutation which results into a dysfunction of the protein leading to the loss of its transcriptional activity associated with a negative effect on the wild type protein in heterozygous status. P53 loss of function mutations have fully been exemplified in the prior art and thus the skilled man in the art can easily identify TP53 mutations (Petitjean A, Mathe E, Kato S, Ishioka C, Tavtigian SV, Hainaut P, Olivier M. Impact of mutant P53 functional properties on TP53 mutation patterns and tumor phenotype: lessons from recent developments in the IARC TP53 database.Hum Mutat. 2007 Jun;28(6):622-9) (http://P53.iarc.fr/). TP53 mutations are mainly missense mutations. Examples of TP53 mutations include but are not limited to R175H, Y163C, C242Y, Y243H, G245S, M246V, R248W, R248Q, R273H, C277Y and C277F. TP 53 mutations are major oncogenic events, strongly associated with the development of many cancers.
As used herein, term “tumoral clone” has its general meaning in the art and refers to a population of tumor cells formed by repeated division from a common cell. The expression “ TP53 mutated tumoral clone” refers to a tumoral clone that harbors at least one TP 53 mutation.
As used herein, the term “MDM2” has its general meaning in the art and refers to E3 ubiquitin-protein ligase Mdm2 P53 binding protein that is a P53-associated protein (Oliner, J. D., et al., Nature 358 (1992) 80-83; Momand, J., et al., Cell 69 (1992) 1237-1245; Chen, J., et al., Mol. Cell. Biol. 13 (1993) 4107-4114; and Bueso-Ramos C. E., et al., Blood 82 (1993) 2617-2623). It is a nuclear phosphoprotein that binds and inhibits transactivation by tumor protein P53, as part of an autoregulatory negative feedback loop. MDM2 also regulates the wild-type P53 protein function by inducing its degradation.
As used herein, the term “MDM2 inhibitor” has its general meaning in the art and refers to therapeutic agents that inhibit the MDM2-P53 interaction. MDM2 inhibitors are usually used as drugs in the field of cancer to stabilize wild-type P53 protein and promote an anti -oncogenic activity. Besides peptides and antibodies, several classes of small-molecule inhibitors with distinct chemical structures have now been reported including, but not limited to, a benzodiazepinedione, a sulphonamide, a chromenotriazolopyrimidine, a morpholinone, a piperidinone, a terphenyl, a chalcone, a pyrazole, an imidazole, an imidazole-indole, an isoindolinone, a pyrrolidinone, a piperidine, a naturally derived prenylated xanthone, a stapled peptide, a benzothi azole, and stictic acid. (Shangary et al., (2008) Ann. Rev. Pharmacol. Toxicol. 49: 223-241). These are derivatives of cis-imidazoline (see e.g. Vassilev et al., (2004) Science 303: 844-848), spiro-oxindole (Ding et al., (2005) J. Am. Chem. Soc. 127: 10130- 10131; Shangary et al., (2008) Proc. Natl. Acad. Sci. USA 105: 3933-3938; Ding et al., (2006) J. Med. Chem. 49: 3432-3435; Shangary et al., (2008) Mol. Cancer Ther. 7: 1533-1542), benzodiazepinedione (Grasberger et al., (2005) J. Med. Chem. 48: 909-912; Parks et al., (2005) Bioorg. Med. Chem. Lett. 15: 765-770; Koblish et al., (2006) Mol. Cancer Ther. 5: 160-169), terphenyl (Yin et al., (2005) Angew. Chem. Int. Ed. Engl. 44: 2704-2707; Chen et al., (2005) Mol. Cancer Ther. 4: 1019-1025), quilinol (Lu, Y., (2006) J. Med. Chem. 49: 3759-3762), chalcone (Stoll et al, (2001) Biochemistry 40: 336-44) and sulfonamide (Galatin et al., (2004) J. Med. Chem. 47: 4163-4165). Examples of MDM2 inhibitors and their mechanism of action and effect on P53 levels are also discussed in Hoe et al., (2014) Nature Revs. 13: 217-236, incorporated herein by reference in its entirety. Methods for identifying MDM2 inhibitors are well known in the art (e.g. Han AR, Durgannavar T, Ahn D, Chung SJ. A FRET-Based Fluorescent Probe to Screen Anticancer Drugs, Inhibiting p73 Binding to MDM2. Chembiochem. 2021 Mar 2;22(5):830-833. doi: 10.1002/cbic.202000660. Epub 2020 Nov 20. PMID: 33103305). For instance, a screening assay using a biosensor involving the physical principle of surface plasm on resonance (SPR) may be performed. For this, P53 may be immobilized on a surface and MDM2 may be put in contact concomitantly with the potential inhibitor. If the potential inhibitor is a MDM2 inhibitor, the percentage of binding between p53 and MDM2 decrease in dose-dependent manner. To investigate the mode of binding of the MDM2 inhibitor, a crystal structure can also be determined.
In some embodiments, the MDM2 inhibitor is a nutlin. As used herein, the term “nutlin” refers to cis-imidazoline analogs that inhibit the interaction between MDM2 and tumor suppressor P53, and which were discovered by screening a chemical library by Vassilev et al., (2004) Science 303: 844-848. Nutlin- 1, Nutlin-2 and Nutlin-3 were all identified in the same screen. However, Nutlin-3, (((±)-4-[4,5-Bis(4-chlorophenyl)-2-(2-isopropoxy-4-methoxy- phenyl)-4,5-dihydro-imidazole-l-carbonyl]-piperazin-2-one) is the compound most commonly used in anti-cancer studies. Inhibiting the interaction between MDM2 and P53 stabilizes P53 and is thought to selectively induce a growth-inhibiting state called senescence in cancer cells. The term “nutlin” as used herein further refers to enantiomers and stereoisomers. The more potent of the two enantiomers, (-)-Nutlin-3 (Nutlin-3 A) ((-)-4-(4,5-Bis(4-chlorophenyl)-2-(2- isopropoxy-4-methoxyphenyl)-4,5-dihydro-lH-imidazole-l-carbonyl)piperazin-2-one can now be synthesized in a highly enantioselective fashion and is arbitrarily referred to as enantiomer because it appears as the first peak from chiral purification of racemic nutlin-3 although its absolute stereocenter assignment is not known. The term “nutlin” may further refer to “second-generation” nutlin” derivatives such as, but not limited to, RG7388 (ChemieTek, Indianapolis, IN) (described by Ding et al. (2013) JMed Chem. 56: 5979-5983 and incorporated herein by reference in its entirety) and to derivatives described in, for example, US Patent Applications 20150211073 and 20170008904.
In some embodiments, the MDM2 inhibitor is Idasanutlin. As used herein, the term “Idasanutlin” has its general meaning in the art and refers to 4-[(2R,3S,4R,5S)-3-(3-chloro-2- fluorophenyl)-4-(4-chloro-2-fluorophenyl)-4-cyano-5-(2,2-dimethylpropyl)pyrrolidine-2- amido]-3-methoxybenzoic acid.
As used herein, the term "treatment" or "therapy" refer to both prophylactic or preventive treatment as well as curative or disease modifying treatment, including treatment of subjects at risk of contracting the disease or suspected to have contracted the disease as well as subjects who are ill or have been diagnosed as suffering from a disease or medical condition, and includes suppression of clinical relapse. The treatment may be administered to a subject having a medical disorder or who ultimately may acquire the disorder, in order to prevent, cure, delay the onset of, reduce the severity of, or ameliorate one or more symptoms of a disorder or recurring disorder, or in order to prolong the survival of a subject beyond that expected in the absence of such treatment. By "therapeutic regimen" is meant the pattern of treatment of an illness, e.g., the pattern of dosing used during therapy. A therapeutic regimen may include an induction regimen and a maintenance regimen. The phrase "induction regimen" or "induction period" refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the initial treatment of a disease. The general goal of an induction regimen is to provide a high level of drug to a subject during the initial period of a treatment regimen. An induction regimen may employ (in part or in whole) a "loading regimen", which may include administering a greater dose of the drug than a physician would employ during a maintenance regimen, administering a drug more frequently than a physician would administer the drug during a maintenance regimen, or both. The phrase "maintenance regimen" or "maintenance period" refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the maintenance of a subject during treatment of an illness, e.g., to keep the subject in remission for long periods of time (months or years). A maintenance regimen may employ continuous therapy (e.g., administering a drug at a regular intervals, e.g., weekly, monthly, yearly, etc.) or intermittent therapy (e.g., interrupted treatment, intermittent treatment, treatment at relapse, or treatment upon achievement of a particular predetermined criteria [e.g., disease manifestation, etc.]). As used herein, the term "risk" in the context of the present invention, relates to the probability that an event will occur over a specific time period and can mean a subject's "absolute" risk or "relative" risk. Absolute risk can be measured with reference to either actual observation post-measurement for the relevant time cohort, or with reference to index values developed from statistically valid historical cohorts that have been followed for the relevant time period. Relative risk refers to the ratio of absolute risks of a subject compared either to the absolute risks of low risk cohorts or an average population risk, which can vary by how clinical risk factors are assessed. Odds ratios, the proportion of positive events to negative events for a given test result, are also commonly used (odds are according to the formula p/(l-p) where p is the probability of event and (1- p) is the probability of no event) to no- conversion. "Risk evaluation," or "evaluation of risk" in the context of the present invention encompasses making a prediction of the probability, odds, or likelihood that an event or disease state may occur, the rate of occurrence of the event or conversion from one disease state to another. Risk evaluation can also comprise prediction of future clinical parameters, traditional laboratory risk factor values, or other indices of relapse, either in absolute or relative terms in reference to a previously measured population. The methods of the present invention may be used to make continuous or categorical measurements of the risk of conversion, thus diagnosing and defining the risk spectrum of a category of subjects defined as being at risk of conversion. In the categorical scenario, the invention can be used to discriminate between normal and other subject cohorts at higher risk.
The method of the present invention is thus particularly suitable for determining whether the patient is at risk of developing a secondary acute leukemia.
As used herein, the term "acute leukemia" means a disease that is characterized by a rapid increase in the numbers of immature blood cells that transform into malignant cells, rapid progression and accumulation of the malignant cells, which spill into the bloodstream and spread to other organs of the body.
Thus the method of the present invention is particularly suitable for determining whether a patient will be eligible for the therapy with the MDM2 inhibitor. The method of the present invention is particularly suitable also for the physician to decide that the patient shall be carefully monitored during and after the treatment so as to select the most accurate clinical decisions. As used herein, the term “hematopoietic stem and progenitor cells” or “HSPCs” refers to blood cells that have the capacity to self-renew and to differentiate into precursors of blood cells. These precursor cells are immature blood cells that cannot self-renew and must differentiate into mature blood cells. HSPCs are typically CD34+ but display a number of phenotypes, such as Lin-CD34+CD38XD90+CD45RA , Lin-CD34+CD38XD90XD45RA Lin-CD34+CD38+IL-3aloCD45RA , and Lin-CD34+CD38+CD10+ (Daley et al., Focus 18:62- 67, 1996; Pimentel, E., Ed., Handbook of Growth Factors Vol. IP: Hematopoietic Growth Factors and Cytokines, pp. 1-2, CRC Press, Boca Raton, Fla., 1994). In some embodiments, the hematopoietic stem and progenitor cells are isolated form peripheral blood cells.
Any culture medium suitable for growth, survival and differentiation of hematopoietic stem and progenitor cells may be used. Typically, it consists of a base medium containing nutrients (a source of carbon, amino acids), a pH buffer and salts, which can be supplemented with serum of human or other origin and/or growth factors and/or antibiotics to which cytokines are added. Typically, the base medium can be RPMI 1640, DMEM, IMDM, X-VIVO or AIM- V medium, all of which are commercially available standard media.
In some embodiments, the culture medium comprises an amount of IL-3 (i.e. interleukin-3), an amount of SCF (i.e. stem cell factor), an amount of Flt-3 ligand (i.e. FMS- like tyrosine kinase 3), an amount of IL-6 (i.e. interleukin-6) and an amount of TPO (i.e. thrombopoietin). The cytokines can be obtained from a variety of sources. They may be purified or recombinant protein and are typically commercially available from different companies, for example R&D Systems or PeproTech.
In some embodiment, the present invention relates to a method of determining whether a patient suffering from a myeloproliferative neoplasm (MPN) is at risk of developing at least one ZP53-mutated tumoral clone under therapy with a MDM2 inhibitor comprising the steps of i) culturing in a culture medium comprising an amount of IL-3 (i.e. interleukin-3), an amount of SCF (i.e. stem cell factor), an amount of Flt-3 ligand (i.e. FMS-like tyrosine kinase 3), an amount of IL-6 (i.e. interleukin-6) and an amount of TPO (i.e. thrombopoietin) a sample of hematopoietic stem and progenitors cells (HSPC) obtained from the patient in presence of an amount of the MDM2 inhibitor for at least 10 days, and ii) detecting the presence and quantifying of ZP53-mutated tumoral clones wherein detection of said clones indicates that the patient is at risk of developing ZP53-mutated tumoral clones and the quantification in comparison with non-treated condition allows to predict the expansion of a pre-existing TP53- mutated clone.
Typically, IL-3 is used in an amount comprised between 40ng/mL and 60ng/mL and is preferably about 50 ng/mL.
Typically, SCF is used in an amount comprised between 80ng/mL and 120ng/mL and is preferably about 100 ng/mL.
Typically, IL-6 is used in an amount comprised between 40ng/mL and 60ng/mL and is preferably about 50 ng/mL.
Typically, Flt-3 ligand is used in an amount comprised between 80ng/mL and 120ng/mL and is preferably about 100 ng/mL.
Typically, TPO is used in an amount comprised between lOng/mL and 30ng/mL and is preferably about 20 ng/mL.
Typically, the MDM2 inhibitor is used in an amount comprised between lOnM and 30nM and is preferably about 20nM.
Typically, in some embodiments, the culture of the hematopoietic stem and progenitor cells is carried out for about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 days. In some embodiments, the culture of the hematopoietic stem and progenitor cells is carried out for at least 10 days.
In some embodiments, detection of the TP53 mutated tumoral clones is performed by any method well known in the art. In some embodiments, the detection of the TP53 mutated tumoral clones is performed by sequencing. Typically, nucleic acids are extracted and purified from the hematopoietic stem and progenitor cells at the end of the culture and sequencing is performed. According to the present invention, a plurality of reads is thus obtained. As used herein, the term “read” refers to a sequence read from a portion of a nucleic acid sample. Typically, a read represents a short sequence of contiguous base pairs in the sample. The read may be represented symbolically by the base pair sequence in A, T, C, and G of the sample portion, together with a probabilistic estimate of the correctness of the base (quality score). As used herein, the term “sequencing” generally means a process for determining the order of nucleotides in a nucleic acid. A variety of methods for sequencing nucleic acids is well known in the art and can be used. In some embodiments, next generation sequencing is carried out. As used herein, the term “next generation sequencing” has its general meaning in the art and refers to sequencing technologies having increased throughput as compared to traditional Sanger- and capillary electrophoresis-based approaches, for example with the ability to generate hundreds of thousands or millions of relatively short sequence reads at a time. Next-generation sequencers are well known in the art and can include a number of different sequencers based on different technologies, such as Illumina (Solexa) sequencing, Roche 454 sequencing, Ion torrent sequencing, SOLiD sequencing, and the like. An example of a sequencing technology that can be used in the present methods is the Illumina platform. The Illumina platform is based on amplification of DNA on a solid surface (e.g., flow cell) using fold-back PCR and anchored primers (e.g., capture oligonucleotides). For sequencing with the Illumina platform, DNA is thus fragmented, and adapters are added to both terminal ends of the fragments (see the preceding step). DNA fragments are attached to the surface of flow cell channels by capturing oligonucleotides which are capable of hybridizing to the adapter ends of the fragments. The DNA fragments are then extended and bridge amplified. After multiple cycles of solid-phase amplification followed by denaturation, an array of millions of spatially immobilized nucleic acid clusters or colonies of single-stranded nucleic acids are generated. Each cluster may include approximately hundreds to a thousand copies of single-stranded DNA molecules of the same template. The Illumina platform uses a sequencing-by-synthesis method where sequencing nucleotides comprising detectable labels (e.g., fluorophores) are added successively to a free 3 'hydroxyl group. After nucleotide incorporation, a laser light of a wavelength specific for the labeled nucleotides can be used to excite the labels. An image is captured and the identity of the nucleotide base is recorded. These steps can be repeated to sequence the rest of the bases. Sequencing according to this technology is described in, for example, U.S. Patent Publication Application Nos. 2011/0009278, 2007/0014362, 2006/0024681, 2006/0292611, and U.S. Pat. Nos. 7,960,120, 7,835,871, 7,232,656, and 7,115,200, each of which is incorporated herein by reference in its entirety.
In some embodiments, the method of the present invention further comprises the steps of i) implementing an algorithm on data that results from the sequencing as to obtain an algorithm output; and ii) determining the probability that the patient will develop at least one TP53 mutated tumoral clone under therapy with the MDM2 inhibitor.
As used herein, the term “algorithm” is any mathematical equation, algorithmic, analytical or programmed process, or statistical technique that takes one or more continuous parameters and calculates an output value, sometimes referred to as an “index” or “index value.” Non-limiting examples of algorithms include sums, ratios, and regression operators, such as coefficients or exponents, biomarker value transformations and normalizations (including, without limitation, those normalization schemes based on clinical parameters, such as gender, age, or ethnicity), rules and guidelines, statistical classification models, and neural networks trained on historical populations. Of particular interest are structural and syntactic statistical classification algorithms, and methods of risk index construction, utilizing pattern recognition features, including established techniques such as cross-correlation, Principal Components Analysis (PCA), factor rotation, Logistic Regression (LogReg), Linear Discriminant Analysis (LDA), Eigengene Linear Discriminant Analysis (ELDA), Support Vector Machines (SVM), Random Forest (RF), Recursive Partitioning Tree (RPART), as well as other related decision tree classification techniques, Shrunken Centroids (SC), StepAIC, Kth-Nearest Neighbor, Boosting, Decision Trees, Neural Networks, Bayesian Networks, Support Vector Machines, and Hidden Markov Models, among others. Other techniques may be used in survival and time to event hazard analysis, including Cox, Weibull, Kaplan-Meier and Greenwood models well known to those of skill in the art.
In some embodiments, the method of the present invention comprises the use of a machine learning algorithm. The machine learning algorithm may comprise a supervised learning algorithm. Examples of supervised learning algorithms may include Average One- Dependence Estimators (AODE), Artificial neural network (e.g., Backpropagation), Bayesian statistics (e.g., Naive Bayes classifier, Bayesian network, Bayesian knowledge base), Case- based reasoning, Decision trees, Inductive logic programming, Gaussian process regression, Group method of data handling (GMDH), Learning Automata, Learning Vector Quantization, Minimum message length (decision trees, decision graphs, etc.), Lazy learning, Instance-based learning Nearest Neighbor Algorithm, Analogical modeling, Probably approximately correct learning (PAC) learning, Ripple down rules, a knowledge acquisition methodology, Symbolic machine learning algorithms, Subsymbolic machine learning algorithms, Support vector machines, Random Forests, Ensembles of classifiers, Bootstrap aggregating (bagging), and Boosting. Supervised learning may comprise ordinal classification such as regression analysis and Information fuzzy networks (IFN). Alternatively, supervised learning methods may comprise statistical classification, such as AODE, Linear classifiers (e.g., Fisher's linear discriminant, Logistic regression, Naive Bayes classifier, Perceptron, and Support vector machine), quadratic classifiers, k-nearest neighbor, Boosting, Decision trees (e.g., C4.5, Random forests), Bayesian networks, and Hidden Markov models. The machine learning algorithms may also comprise an unsupervised learning algorithm. Examples of unsupervised learning algorithms may include artificial neural network, Data clustering, Expectation- maximization algorithm, Self-organizing map, Radial basis function network, Vector Quantization, Generative topographic map, Information bottleneck method, and IBSEAD. Unsupervised learning may also comprise association rule learning algorithms such as Apriori algorithm, Eclat algorithm and FP-growth algorithm. Hierarchical clustering, such as Single linkage clustering and Conceptual clustering, may also be used. Alternatively, unsupervised learning may comprise partitional clustering such as K-means algorithm and Fuzzy clustering. In some instances, the machine learning algorithms comprise a reinforcement learning algorithm Examples of reinforcement learning algorithms include, but are not limited to, temporal difference learning, Q-learning and Learning Automata. Alternatively, the machine learning algorithm may comprise Data Pre-processing.
In some embodiments, the output obtained by the algorithm is a score. As used herein, the term “score” refers to a piece of information, usually a number that conveys the result of the subject on a test. A risk scoring system separates a patient population into different risk groups; herein the process of risk stratification classifies the patients into very high-risk, high- risk, intermediate-risk and low-risk groups.
A further object of the present invention relates to a kit or device for performing the method of the present invention, comprising means for carrying out the culture and detecting the TP53 mutated tumoral clones as described above. In some embodiments, the kits or devices of the present invention comprise at least one sample collection container for sample collection. Collection devices and container include but are not limited to syringes, lancets, collection tubes. In some embodiments, the container contains the predetermined amount of the different cytokines (e.g. IL-3, SCF, IL-6, FLT-3 Ligand and TPO) and the MDM2 inhibitor. In some embodiments, the kits or devices described herein further comprise instructions for using the kit or device and interpretation of results. In some embodiments, the kit or device of the present invention further comprises a microprocessor to implement an algorithm so as to determine the probability that the patient will develop TP53 mutated tumoral clones. In some embodiments, the kit or device of the present invention further comprises a visual display and/or audible signal that indicates the probability determined by the microprocessor.
The invention will be further illustrated by the following figures and examples. However, these examples and figures should not be interpreted in any way as limiting the scope of the present invention.
FIGURES:
Figure 1: Increase of mutant TP53 VAFs in HSPC of 7P5.?-mutated MPN patients upon MDM2 inhibitor treatment. (A) Comparison of various TP53 mutations VAFs in CD34+ cells of MPN1 patient after 10 days with or without 20 nM of idasanutlin. (B) Comparison of all TP 53 mutations VAFs in CD34+ cells of 8 different MPN patients after 10 days in presence or absence of idasanutlin 20 nM. (C) Mean +/- SD of VAFs for all TP 53 mutants found in 8 MPN patients after 10 days in presence or absence of idasanutlin 20 nM or Ruxolitinib 70nM in 4 other MPN patients. A paired t-test was used, ** p < 0.01, ns: non significant Figure 2: Absence of variation of non TP53 mutations in HSPC of 7P5J-mutated MPN patients upon MDM2 inhibitor treatment. Comparison of VAFs for mutations found in other genes in CD34 positive cells of the same MPN patients. NT: Not treated. **: p < 0.01 using a Student test for statistical comparison.
EXAMPLE:
Material and methods
Patients. MPN patients were selected on the basis of having one driver mutation and at least one TP53 mutation detectable by NGS. Most of these patients also harbored mutations in other genes. Clinical and molecular characteristics are given in Table 1.
In vitro assay CD34+ cells from MPN patients after NGS analysis were prospectively cryopreserved. Peripheral mononuclear cells were isolated from whole blood using Ficoll (Eurobio) and CD34+ cells sorted using a column-free immunomagnetic approach (EasySep®, StemCell). 105 CD34+ cells were cultured in CTS StemPro™ HSC expansion medium (Thermo Fisher) in which the following cytokines were added: IL-3 (50 ng/mL), SCF (100 ng/mL), TPO (20 ng/mL), IL-6 (50 ng/mL), FLT-3 Ligand (100 ng/mL). Idasanutlin (provided by Roche) was added at 20 nM. After 10 days of culture, cells were harvested, DNA extracted and submitted to NGS analysis.
NGS analysis. We used a capture-based custom NGS panel (Sophia Genetics) targeting 36 myeloid genes (ABU; ASXL1; BRAF; CALR; CBL; CCND2; CEBPA; CSF3R; CUX1; DNMT3A; ETNK1; ETV6; EZH2; FLT3; HRAS; IDH1; IDH2; IKZF1; JAK2; KIT; KRAS; MPL; NFE2; NPM1; NRAS; PTPN11; RUNX1; SETBP1; SF3B1; SH2B3; SRSF2; TET2; TP53; U2AF1; WT1; ZRSR2). Libraries were prepared on 200ng extracted from whole blood DNA (Qiagen) and sequencing was performed on a MiSeq® instrument (Illumina). Bioinformatics were performed at Sophia Genetics (Switzerland) using the SOPHIA DDM software and significant variants were retained with a sensitivity of 1%.
Single cell analysis. The same sample cultured in vitro was used for single cell analysis. CD34+ were resuspended in Tapestri® cell buffer (Mission Bio) and quantified using an automatic cell counter (Biorad). Viable single cells (3,000-4,000 cells/mΐ) were encapsulated using a Tapestri® microfluidics cartridge (Mission Bio), lysed, and barcoded. Barcoded samples were then subjected to targeted PCR amplification of a custom 96 amplicons covering 17 genes known to be mutated in MPN (. ASXL1 ; CALR; CBL; DNMT3A; EZH2; IDHF, IDH2, JAK2; KRAS; MPL; NFE2; NRAS; SF3B1; SRSF2; TET2; TP53; U2AF1). PCR products were removed from individual droplets, purified with Ampure XP beads (Beckman Coulter), and used as a template for PCR to incorporate Illumina i5/i7 indices. PCR products were purified a second time, quantified via an Agilent Bioanalyzer and pooled to be sequenced. Library pools were sequenced on aNextSeq instrument (Illumina). The Tapestri Insights® pipeline was used to filter variants (data not shown) and samples were included if they harbored three or more protein-encoding, non-synonymous/insertion/deletion variants and more than 1000 cells with definitive genotype for all protein-coding variants within the sample. We next sought to define genetic clones, which we identified as cells that possessed identical genotype calls for the protein-encoding variants of interest. Importantly, almost 95% of the panel amplicons in each sample had sufficient coverage to annotate variants. The estimated median allele dropout (ADO) rate was 8.74% (IQR: 6.5% - 10.6%, data not shown). From the variants annotated for each sample, we first removed those with low quality (<20% in the Tapestri Insights® software) and low frequency (< 1% cells) (data not shown). As a result, we detected a total of 37 different variants passing the pre-filtering step across the 8 patients (data not shown) on the NGS analysis reference, with a median of 4 variants per patient (IQR: 3.25 - 4.75). All variants detected in NGS analysis below the 1% threshold were not taken into account in the single cell analysis. Study approval. The study was accepted by the local IRB (Comite d’Evaluation de TEthique des projets de Recherche Biomedicale (CEERB) Paris Nord) and patients have given informed consent for the study.
Results
Direct and rapid selection of 7P5.?-mutated clones under MDM2 treatment
After the observation of increased allelic frequencies of TP53 mutations in 5 patients treated in vivo with idasanutlin, an MDM2 inhibitor, we addressed the question of a direct effect of this drug on the selection of mutated cells. We aimed at reproducing the clinical observation with a short-term in vitro culture of patient samples using a controlled medium to support the hypothesis of a direct targeted cellular effect of MDM2 inhibition. Eight patients harboring at least the presence of one driver mutation and one TP53 mutation were examined for the impact of idasanutlin on the selection of ZP53-mutated subclones. Patient’ clinical and molecular characteristics are depicted in Table 1. We first determined the optimal culture conditions and idasanutlin concentrations that limited cellular proliferation of CD34+ cells from 3 MPN patients (data not shown). A dose of 20nM idasanutlin was identified as optimal and used in the subsequent experiments.
CD34+ cells from ZP53-mutated patients were cultured with or without idasanutlin. After 10 days the cells were retrieved and NGS sequenced as previously described (19). To validate the culture conditions, we compared the allelic frequencies (VAF) of every mutation found after 10 days without treatment to those found in whole blood DNA. For a total of 37 mutations, we observed an excellent correlation between VAFs measured by NGS after culture of CD34+ cells and VAFs measured in bulk whole blood DNA (R=0.979, p< 0.0001) (data not shown) demonstrating that the culture conditions reproduced the in vivo situation without altering the molecular architecture of MPN cells. To estimate the accuracy of this assay for reproducing the in vivo selection of ZP53-mutated clones, we analyzed cells taken from a patient (#MPN1) who had experienced an expansion of TP 53 mutant clones in vivo during idasanutlin therapy (18) and found a similar increase of TP53 mutation VAFs in CD34+ cells in vitro (Figure 1A). We concluded the in vitro culture conditions we defined closely replicated the in vivo process of a clonal selection of TP53 mutated cells. We then tested 7 additional patient samples using this assay and observed an increase of each TP53 mutant VAF after idasanutlin treatment compared to the untreated (NT) conditions (Figure IB). In patients with several TP53 mutations, the VAF of each of these mutations increased independently. Altogether, the mean VAF of TP53 mutations was significantly higher in presence of the MDM2 inhibitor treatment compared to the untreated condition (7.38%, us 3.34% respectively, p < 0.01) (Figure 1C). Importantly, such increase was not observed under in vitro exposure to ruxolitinib (Figure 1C). To evaluate the specificity of the selection for TP53 mutations, we considered in each sample the variants found in all genes. Contrary to TP53 mutations, the VAF of mutations found in other genes showed no significant variation after culture with idasanutlin (driver mutations, NT: 38,29% vs Idasanutlin: 39,93%, p=0,25, additional non-TP53 mutations, NT : 11,59% vs idasanutlin: 11,98%, p=0,30, strongly indicating a specific selection of the TP53 mutation-bearing cells by idasanutlin (Figure 2).
In four of the eight patients tested, we identified TP53 mutations in the treated sample that were not detected in the untreated sample or in the whole blood DNA. We tracked these emerging mutations by carefully checking the raw sequencing data from whole blood using IGV software and identified in each case a significant amount of mutant reads (data not shown). For patient #MPN1 who received idasanutlin therapy, the presence of these low level mutations was further confirmed by their detection in a subsequent blood sample taken 3 months later. In patient #MPN4 treated only with hydroxyurea for 20 years, the mutation (p.Y176C) detected after in vitro culture using idasanutlin was eventually found in a follow-up whole blood sample taken one year later. This observation confirmed prior experiments suggesting that idasanutlin is not likely to induce TP53 mutations but rather participates in their selection (18).
Altogether, using this new in vitro culture assay with defined and controlled medium combined with NGS analysis after 10 days of culture, we confirmed in 8 different patients and for a total of 16 TP 53 mutations a direct and rapid effect of idasanutlin exposure on the selection of ZP53-mutated MPN CD34+ cells. Importantly, this test appears able to uncover TP53- mutated clones that may be selected in vivo before initiating treatment with an MDM2 inhibitor, even when they are undetectable in patients’ blood using NGS.
Single cell-derived clonal architecture and selection of TP53-mutant MPN cells
Both driver and non-driver mutations found in MPN cells target important regulatory pathways such as intracellular signaling, DNA methylation, histone modifications or splicing processes that could modulate the effects of drugs like MDM2 inhibitors. To better decipher the impact of MDM2 inhibition on clonal selection we next investigated the co-occurrence and order of mutation acquisition for each of the 8 patients tested in vitro. Clonal architecture was determined using the single-cell high-throughput microfluidic Tapestri platform® coupled to the Tapestri Insights® bioinformatics software (Mission Bio). As recently reported (25) this approach permitted accurate analysis of thousands of single cells per specimen. From 8 patient samples a total of 45,443 cells were sequenced with a median of 4,923 cells per sample (interquartile range [IQR]: 3820 - 8172). The panel designed for this study covered 17 of the most frequent MPN mutated genes. The median sequencing coverage was 100 reads per amplicon per cell (IQR: 48.07 - 127.2). To validate the accuracy of the variant identification in single cells, we compared the allelic frequency of each mutation found with single cell analysis with the VAFs found with NGS of whole blood DNA. Among 37 variants identified by whole blood NGS and covered by the panel, 100% (33 SNV and 4 indels) were also detected using the single-cell approach. For each variant there was an excellent correlation between the proportions of mutant alleles detected at single-cell level and the VAFs measured with whole blood DNA (data not shown). When considering all the variants together, the allelic fractions quantified by single-cell and whole blood NGS were highly correlated (R=0.991, p< 0.0001, data not shown).
The results of the single cell analysis allowed us to infer the order of acquisition of somatic events and hence reconstitute the clonal architecture for each patient (data not shown).
In five patients we observed a major clone harboring 1-3 variants and comprising 32% to 75% of the cells associated with 2 to 9 minor subclones with less than 10% of the cells (#MPN1 to 5, data not shown). In three patients several clones of comparable size were present without one dominant clone (#MPN6 to 8) (data not shown). In most cases the first molecular event was a driver mutation in JAK2 or CALR. Only in #MPN2 and #MPN3 patients were DNMT3A and U2AF1 mutations, respectively, the initial events (data not shown).
Several striking observations on the clonal architecture of MPN rapidly emerged from this study. First, clonal evolution of MPN was rarely linear with sequential accumulation of mutations in the same clone. To the contrary, we observed branching evolution with several clones evolving simultaneously in the majority of samples (data not shown). Another important finding was that JAK2V617F homozygous subclones were present in all JAK2,¾/7/- mutated patients evaluated. In #MPN1 patient, homologous recombination giving rise to a homozygous clone occurred early in disease development and was present in almost all the single cells tested. In other patients, homologous recombination occurred as a late event and was present in a minor subclone (#MPN3), or even in several minor subclones (#MPN8). Finally, we observed that TP53 mutations always appeared late during clonal evolution, and exclusively in clones harboring a driver mutation (JAK2 or CALR in this study). Although the number of patients tested was limited, it is unlikely that these conclusions can be attributed to chance as this association was observed in 8/8 patients. Furthermore, we tested several patients with multiple TP53 mutations which all developed in distinct clones, but always in cells carrying the driver mutation. For example, in #MPN1 a major JAK2IASXL1 mutated clone was present from which emerged three subclones, each with a different TP53 mutation (data not shown). Multiple independent ZP53-mutated subclones within the driver clone were also detected in #MPN3 and #MPN4 patients (data not shown). The absence of JAK2 wild- type/ TP 53 mutated cells was unexpected since JAK2V617F positive MPN may evolve to JAK2 wild-type / TP53 mutated acute leukemia while genotypic reversion by mitotic recombination or gene deletion has been previously excluded as an explanation for the development of JAK2 wild-type leukemia in JAK2 -mutated patients (22).
Interestingly, several patients had additional and independent clones with mutations affecting epigenetic genes ( TET2 and DNMT3A ) in small numbers of cells (#MPN5, 6 and 7, data not shown). According to the advanced age of these patients (83, 82 and 67 years), these clones could simply reflect clonal hematopoiesis of indeterminate potential (CHIP) without oncogenic potential occurring independently of MPN-related clones.
The concomitant analysis on the same samples of MDM2 inhibitor treatment and single cell DNA clonal architecture allowed us to determine the molecular background in which TP53 mutations are acquired at the single cell level, and whether this background may affect their selection by MDM2 inhibitors. We found that TP53 mutations may be acquired in three distinct scenarios: either in a clone mutated for a driver gene and an epigenetic gene (TET2, DNMT3A , ASXL1 or EZH2), which is the most frequent situation (#MPN1, 2, 6), or in a clone mutated for a driver gene and a splicing factor (U2AF1 in #MPN3) or in a clone with a driver mutation only that has undergone homologous recombination (#MPN7) or not (#MPN4, 5, 8). Thus, TP53 mutations may be acquired in a large array of molecular landscapes without evidence for a specific association with a particular pathway alteration, with the notable exception of the presence of driver mutations. With idasanutlin treatment in vitro , we observed a significant increase in the VAFs of every ZP53-mutated clone. We did not identify any additional mutations that might favor or antagonize idasanutlin-dependent selection of ZP53-mutated clones, but numbers of clones were relatively small and confirmatory studies would be of interest. Nevertheless, our study shows that the selection by idasanutlin at the single cell level was due to the presence of TP53 mutation itself, independently of associated mutations.
Discussion
In the aim of preventing leukemic transformation, understanding which circumstances may favor the selection of cells carrying TP53 mutations is crucial. MPN patients are treated for years with various anti-proliferative drugs, some treatments have been clearly shown to favor the transformation process (23). However, the participation of long-term treatment in MPN patients in the clonal selection of TP53 mutations is still elusive. Particularly, hydroxyurea has been suspected but no definitive correlation has been established (5). Recent anti-cancer therapeutic approaches aim at enhancing the p53 tumor suppressor activity, notably using MDM2 inhibitors such as nutlins (15) to rescue p53 from degradation (24). This novel class of drugs is now tested in a wide variety of cancers including hematological malignancies.
Several parameters may influence the clonal evolution in vivo and to demonstrate a direct effect of idasanutlin in the expansion of ZP53-mutated clones we performed in vitro culture of patients-derived HSPCs in a defined medium. We observed after a short delay (10 days) a selection of ZP53-mutated cells at the expense of wild-type cells. Importantly, this was reproducible in 8 out of 8 patients, with 16 different TP 53 mutations targeting the DNA binding domain between exon 5 and 8 (five mutations in exon 5, four mutations in exon 6, five in exon 7 and two in exon 8). In patients harboring several TP53 mutations, we observed that each individual mutation VAF increased. In the same time, none of the VAF of other mutations present in these patients increased. This demonstrated that MDM2 inhibitor directly favors the expansion of cells harboring TP53 mutations, an effect not observed in this study with JAK inhibition using ruxolitinib, and raises the question of the mechanism by which the inhibition of MDM2 acts on clonal selection. One explanation is that idasanutlin treatment activates the p53 pathway and cell death in wild-type p53 cells, which would not occur in p53 mutant cells with loss of wild-type functions. The other explanation is that mutations confer gain of function mutations to p53 leading to positive selection of mutated cells. The possibility of gain of function mutations in p53 is still debated but it would be interesting to test the drug on TP53- mutated or inactivated cells to explore a possible gain of function of the mutated forms in the survival of these cells under treatment. Thus, we developed an in vitro assay using patient’ cells which has the potential to uncover ZP53-mutated clones that were not detected by NGS. The results presented herein establish for the first time the causality of this treatment in the selection of MPN sub-clones with a high potential for leukemic transformation. We developed an in vitro assay that may help identify patients with ZP53-mutated clones undetectable with standard NGS before starting an MDM2 inhibitor. With the exception of driver mutations, we did not identify any gene or pathway being redundantly mutated and co segregating with the TP53 mutations, suggesting that only TP53 mutations confer a selective advantage under idasanutlin therapy. Therefore, one can suspect that selection of TP53 mutated cells may occur whatever the tissue and the subtype of cancer.
Tables:
Figure imgf000022_0001
Table 1: Clinical and molecular characteristics of patients harboring at least the presence of one driver mutation and one TP53 mutation. REFERENCES:
Throughout this application, various references describe the state of the art to which this invention pertains. The disclosures of these references are hereby incorporated by reference into the present disclosure. 1. Vannucchi AM et al. Mutations and prognosis in primary myelofibrosis. Leukemia 2013 ;27(9): 1861-1869.
2. Guglielmelli P et al. MIPSS70: Mutation-Enhanced International Prognostic Score System for Transplantation-Age Patients With Primary Myelofibrosis. J. Clin. Oncol. 2018;36(4):310- 318.
3. Tefferi A et al. CALR and ASXL1 mutations-based molecular prognostication in primary myelofibrosis: an international study of 570 patients. Leukemia 2014;28(7): 1494-1500.
4. Tefferi A et al. Targeted deep sequencing in polycythemia vera and essential thrombocythemia. Blood Adv 2016; 1(1):21— 30.
5. Kubesova B et al. Low-burden TP53 mutations in chronic phase of myeloproliferative neoplasms: association with age, hydroxyurea administration, disease type and JAK2 mutational status. Leukemia 2018;32(2):450-461.
6. Grinfeld J et al. Classification and Personalized Prognosis in Myeloproliferative Neoplasms. N. Engl. J. Med. 2018;379(15): 1416-1430.
7. Lane DP. Cancer. p53, guardian of the genome. Nature 1992;358(6381): 15-16.
8. Rivlin N, Koifman G, Rotter V. p53 orchestrates between normal differentiation and cancer. Semin Cancer Biol 2015;32:10-17.
9. Muller PAJ, Vousden KH. p53 mutations in cancer. Nat Cell Biol 2013;15(1): 2—8.
10. Loh SN. Follow the Mutations: Toward Class-Specific, Small-Molecule Reactivation of p53. Biomolecules 2020;10(2). doi:10.3390/bioml0020303
11. Maslah N et al. Synergistic effects of PRIMA-lMet (APR-246) and 5-azacitidine in TP53- mutated myelodysplastic syndromes and acute myeloid leukemia. Haematologica 2020; 105(6): 1539-1551.
12. Kubbutat MH, Jones SN, Vousden KH. Regulation of p53 stability by Mdm2. Nature 1997;387(6630):299-303.
13. Honda R, Tanaka H, Yasuda H. Oncoprotein MDM2 is a ubiquitin ligase E3 for tumor suppressor p53. FEBS Lett 1997;420(l):25-27.
14. Lukashchuk N, Vousden KH. Ubiquitination and degradation of mutant p53. Mol Cell Biol 2007;27(23): 8284-8295. 15. Vassilev LT et al. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science 2004;303(5659):844-848.
16. Lu M et al. Combination treatment in vitro with Nutlin, a small-molecule antagonist of MDM2, and pegylated interferon-a 2a specifically targets JAK2V617F -positive polycythemia vera cells. b/ooί/2012;120(15):3098-3105.
17. Mascarenhas J et al. Oral idasanutlin in patients with polycythemia vera. Blood 2019; 134(6): 525-533.
18. Marcellino BK et al. Transient expansion of TP53 mutated clones in polycythemia vera patients treated with idasanutlin. Blood Adv 2020;4(22): 5735-5744.
19. Cassinat B et al. CCND2 mutations are infrequent events in BCR-ABL1 negative myeloproliferative neoplasm patients. Haematologica 2021;106(3):863-864.
20. Lasho TL et al. Targeted next-generation sequencing in blast phase myeloproliferative neoplasms. Blood Adv 2018;2(4):370-380.
21. Luque Paz D et al. Leukemic evolution of polycythemia vera and essential thrombocythemia: genomic profiles predict time to transformation. Blood Adv 2020;4(19):4887-4897.
22. Beer PA et al. Two routes to leukemic transformation after a JAK2 mutation-positive myeloproliferative neoplasm. Blood 2010; 115(14):2891-2900.
23. Kiladjian J-J, Chevret S, Dosquet C, Chomienne C, Rain J-D. Treatment of polycythemia vera with hydroxyurea and pipobroman: final results of a randomized trial initiated in 1980. J. Clin. Oncol. 2011;29(29):3907-3913.
24. Duffy MJ, Synnott NC, O’Grady S, Crown J. Targeting p53 for the treatment of cancer. Semin Cancer Biol [published online ahead of print: July 31, 2020]; doi:10.1016/j.semcancer.2020.07.005
25. Miles LA, Bowman RL, Merlinsky TR, et al. Single-cell mutation analysis of clonal evolution in myeloid malignancies. Nature. 2020;587(7834):477-482.

Claims

CLAIMS:
1. A method of determining whether a patient suffering from a myeloproliferative neoplasm (MPN) is at risk of developing at least one ZP53-mutated tumoral clone under therapy with a MDM2 inhibitor comprising the steps of i) culturing a sample of hematopoietic stem and progenitors cells (HSPC) obtained from the patient in presence of an amount of the MDM2 inhibitor for at least 5 days, and ii) detecting the presence of ZP53-mutated tumoral clones wherein detection of said clones indicates that the patient is at risk of developing ZP53-mutated tumoral clones.
2. The method of claim 1 wherein the patient harbours at least one mutation in JAK2, MPL, or CALR.
3. The method of clam 1 wherein the MDM2 inhibitor is a nutlin.
4. The method of claim 1 wherein the MDM2 inhibitor is Idasanutlin.
5. The method of claim 1 wherein the culture medium comprises an amount of IL-3 (i.e. interleukin-3), an amount of SCF (i.e. stem cell factor), an amount of Flt-3 ligand (i.e. FMS-like tyrosine kinase 3), an amount of IL-6 (i.e. interleukin-6) and an amount of TPO (i.e. thrombopoietin).
6. The method of claim 1 wherein the culture of the hematopoietic stem and progenitor cells is carried out for about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 days.
7. The method of claim 1 wherein the detection of the ZP53-mutated tumoral clones is performed by sequencing.
8. The method of claim 1 which further comprises the steps of i) implementing an algorithm on data that results from the sequencing as to obtain an algorithm output; and ii) determining the probability that the patient will develop at least one ZP53-mutated tumoral clone under therapy with the MDM2 inhibitor.
9. A kit or device for performing the method according to claim 1 to 9, comprising means for carrying out the culture and detecting the ZP53-mutated tumoral clones.
PCT/EP2022/060039 2021-04-15 2022-04-14 In vitro tests allowing to identify the potential for mdm2 inhibitors to induce the selection of mutations in patients suffering from a myeloproliferative neoplasm WO2022219128A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP21305499 2021-04-15
EP21305499.2 2021-04-15

Publications (1)

Publication Number Publication Date
WO2022219128A1 true WO2022219128A1 (en) 2022-10-20

Family

ID=75801527

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2022/060039 WO2022219128A1 (en) 2021-04-15 2022-04-14 In vitro tests allowing to identify the potential for mdm2 inhibitors to induce the selection of mutations in patients suffering from a myeloproliferative neoplasm

Country Status (1)

Country Link
WO (1) WO2022219128A1 (en)

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060024681A1 (en) 2003-10-31 2006-02-02 Agencourt Bioscience Corporation Methods for producing a paired tag from a nucleic acid sequence and methods of use thereof
US7115200B2 (en) 2002-05-24 2006-10-03 Mayfran International B. V. Device for receiving and separating chips created by machine-tools and coolant (drive)
US20060292611A1 (en) 2005-06-06 2006-12-28 Jan Berka Paired end sequencing
US20070014362A1 (en) 2005-07-15 2007-01-18 Cruz Diego S Method and apparatus for motion compensated temporal filtering
US7232656B2 (en) 1998-07-30 2007-06-19 Solexa Ltd. Arrayed biomolecules and their use in sequencing
US7835871B2 (en) 2007-01-26 2010-11-16 Illumina, Inc. Nucleic acid sequencing system and method
US7960120B2 (en) 2006-10-06 2011-06-14 Illumina Cambridge Ltd. Method for pair-wise sequencing a plurality of double stranded target polynucleotides
US20150211073A1 (en) 2013-12-06 2015-07-30 Hoffmann-La Roche Inc. Mrna-based gene expression for personalizing patient cancer therapy with an mdm2 antagonist
US20170008904A1 (en) 2015-07-10 2017-01-12 Arvinas, Inc. Mdm2-based modulators of proteolysis and associated methods of use
WO2019213074A1 (en) * 2018-04-30 2019-11-07 Kartos Therapeutics, Inc. Methods of treating cancer

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7232656B2 (en) 1998-07-30 2007-06-19 Solexa Ltd. Arrayed biomolecules and their use in sequencing
US7115200B2 (en) 2002-05-24 2006-10-03 Mayfran International B. V. Device for receiving and separating chips created by machine-tools and coolant (drive)
US20060024681A1 (en) 2003-10-31 2006-02-02 Agencourt Bioscience Corporation Methods for producing a paired tag from a nucleic acid sequence and methods of use thereof
US20060292611A1 (en) 2005-06-06 2006-12-28 Jan Berka Paired end sequencing
US20070014362A1 (en) 2005-07-15 2007-01-18 Cruz Diego S Method and apparatus for motion compensated temporal filtering
US7960120B2 (en) 2006-10-06 2011-06-14 Illumina Cambridge Ltd. Method for pair-wise sequencing a plurality of double stranded target polynucleotides
US7835871B2 (en) 2007-01-26 2010-11-16 Illumina, Inc. Nucleic acid sequencing system and method
US20110009278A1 (en) 2007-01-26 2011-01-13 Illumina, Inc. Nucleic acid sequencing system and method
US20150211073A1 (en) 2013-12-06 2015-07-30 Hoffmann-La Roche Inc. Mrna-based gene expression for personalizing patient cancer therapy with an mdm2 antagonist
US20170008904A1 (en) 2015-07-10 2017-01-12 Arvinas, Inc. Mdm2-based modulators of proteolysis and associated methods of use
WO2019213074A1 (en) * 2018-04-30 2019-11-07 Kartos Therapeutics, Inc. Methods of treating cancer

Non-Patent Citations (51)

* Cited by examiner, † Cited by third party
Title
"Handbook of Growth Factors Vol. III: Hematopoietic Growth Factors and Cytokines", 1994, CRC PRESS, pages: 1 - 2
B KUBESOVA ET AL: "Low-burden TP53 mutations in chronic phase of myeloproliferative neoplasms: association with age, hydroxyurea administration, disease type and JAK2 mutational status", LEUKEMIA, vol. 32, no. 2, 24 July 2017 (2017-07-24), London, pages 450 - 461, XP055719410, ISSN: 0887-6924, DOI: 10.1038/leu.2017.230 *
BEER PA ET AL.: "Two routes to leukemic transformation after a JAK2 mutation-positive myeloproliferative neoplasm", BLOOD, vol. 115, no. 14, 2010, pages 2891 - 2900
BUESO-RAMOS C. E. ET AL., BLOOD, vol. 82, 1993, pages 2617 - 2623
CASSINAT B ET AL.: "CCND2 mutations are infrequent events in BCR-ABL1 negative myeloproliferative neoplasm patients", HAEMATOLOGICA, vol. 106, no. 3, 2021, pages 863 - 864
CHEN ET AL., MOL. CANCER THER., vol. 4, 2005, pages 1019 - 1025
CHEN, J. ET AL., MOL. CELL. BIOL., vol. 13, 1993, pages 4107 - 4114
DALEY ET AL., FOCUS, vol. 18, 1996, pages 62 - 67
DING ET AL., J. AM. CHEM. SOC., vol. 127, 2005, pages 10130 - 10131
DING ET AL., J. MED. CHEM., vol. 49, 2006, pages 3759 - 3762
DING ET AL., JMED CHEM, vol. 56, 2013, pages 5979 - 5983
DUFFY MJSYNNOTT NCO'GRADY SCROWN J: "Targeting p53 for the treatment of cancer", SEMIN CANCER BIOL, 31 July 2020 (2020-07-31)
GALATIN ET AL., J. MED. CHEM., vol. 47, 2004, pages 4163 - 4165
GRASBERGER ET AL., J. MED. CHEM., vol. 48, 2005, pages 909 - 912
GUGLIELMELLI P ET AL.: "MIPSS70: Mutation-Enhanced International Prognostic Score System for Transplantation-Age Patients With Primary Myelofibrosis", J. CLIN. ONCOL., vol. 36, no. 4, 2018, pages 310 - 318
HAN ARDURGANNAVAR TAHN DCHUNG SJ: "A FRET-Based Fluorescent Probe to Screen Anticancer Drugs, Inhibiting p73 Binding to MDM2", CHEMBIOCHEM, vol. 22, no. 5, 20 November 2020 (2020-11-20), pages 830 - 833
HOE ET AL., NATURE REVS, vol. 13, 2014, pages 217 - 236
HONDA RTANAKA HYASUDA H: "Oncoprotein MDM2 is a ubiquitin ligase E3 for tumor suppressor p53", FEBSLETT, vol. 420, 1997, pages 25 - 27, XP004261663, DOI: 10.1016/S0014-5793(97)01480-4
JOONIL JUNG ET AL: "TP53 mutations emerge with HDM2 inhibitor SAR405838 treatment in de-differentiated liposarcoma", NATURE COMMUNICATIONS, vol. 7, 31 August 2016 (2016-08-31), pages 12609, XP055522056, DOI: 10.1038/ncomms12609 *
KILADJIAN J-JCHEVRET SDOSQUET CCHOMIENNE CRAIN J-D: "Treatment of polycythemia vera with hydroxyurea and pipobroman: final results of a randomized trial initiated in 1980", J. CLIN. ONCOL., vol. 29, no. 29, 2011, pages 3907 - 3913
KOBLISH ET AL., MOL. CANCER THER., vol. 5, 2006, pages 160 - 169
KUBBUTAT MHJONES SNVOUSDEN KH: "Regulation of p53 stability by Mdm2", NATURE, vol. 387, no. 6630, 1997, pages 299 - 303, XP002108339, DOI: 10.1038/387299a0
KUBESOVA B ET AL.: "Low-burden TP53 mutations in chronic phase of myeloproliferative neoplasms: association with age, hydroxyurea administration, disease type and JAK2 mutational status", LEUKEMIA, vol. 32, no. 2, 2018, pages 450 - 461, XP055719410, DOI: 10.1038/leu.2017.230
LASHO TL ET AL.: "Targeted next-generation sequencing in blast phase myeloproliferative neoplasms", BLOODADV, vol. 2, no. 4, 2018, pages 370 - 380
LOH SN: "Follow the Mutations: Toward Class-Specific, Small-Molecule Reactivation of p53", BIOMOLECULES, vol. 10, 2020
LU M ET AL.: "Combination treatment in vitro with Nutlin, a small-molecule antagonist of MDM2, and pegylated interferon-a 2a specifically targets JAK2V617F-positive polycythemia vera cells", BLOOD, vol. 120, no. 15, 2012, pages 3098 - 3105, XP055719377, DOI: 10.1182/blood-2012-
LUKASHCHUK NVOUSDEN KH: "Ubiquitination and degradation of mutant p53", MOL CELL BIOL, vol. 27, no. 23, 2007, pages 8284 - 8295
LUQUE PAZ D ET AL.: "Leukemic evolution of polycythemia vera and essential thrombocythemia: genomic profiles predict time to transformation", BLOOD ADV, vol. 4, no. 19, 2020, pages 4887 - 4897
MARCELLINO BK ET AL.: "Transient expansion of TP53 mutated clones in polycythemia vera patients treated with idasanutlin", BLOODADV, vol. 4, no. 22, 2020, pages 5735 - 5744, XP055837159, DOI: 10.1182/bloodadvances.2020002379
MARCELLINO BRIDGET K. ET AL: "Transient expansion of TP53 mutated clones in polycythemia vera patients treated with idasanutlin", vol. 4, no. 22, 24 November 2020 (2020-11-24), pages 5735 - 5744, XP055837159, ISSN: 2473-9529, Retrieved from the Internet <URL:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7686898/pdf/advancesADV2020002379.pdf> DOI: 10.1182/bloodadvances.2020002379 *
MASCARENHAS J ET AL.: "Oral idasanutlin in patients with polycythemia vera", BLOOD, vol. 134, no. 6, 2019, pages 525 - 533, XP086699270, DOI: 10.1182/blood.2018893545
MASLAH N ET AL.: "Synergistic effects of PRIMA-lMet (APR-246) and 5-azacitidine in TP53-mutated myelodysplastic syndromes and acute myeloid leukemia", HAEMATOLOGICA, vol. 105, no. 6, 2020, pages 1539 - 1551
MILES LABOWMAN RLMERLINSKY TR ET AL.: "Single-cell mutation analysis of clonal evolution in myeloid malignancies", NATURE, vol. 587, no. 7834, 2020, pages 477 - 482, XP037298653, DOI: 10.1038/s41586-020-2864-x
MIN LU ET AL: "Combination treatment in vitro with Nutlin, a small-molecule antagonist of MDM2, and pegylated interferon-_ 2a specifically targets JAK2V617F-positive polycythemia vera cells", BLOOD, 1 January 2012 (2012-01-01), pages 3098 - 3105, XP055719377, Retrieved from the Internet <URL:https://ashpublications.org/blood/article-pdf/120/15/3098/1359739/zh804112003098.pdf> DOI: 10.1182/blood-2012- *
MOMAND, J. ET AL., CELL, vol. 69, 1992, pages 1237 - 1245
MORHAYIM JESS ET AL: "Identification of osteolineage cell-derived extracellular vesicle cargo implicated in hematopoietic support", vol. 34, no. 4, 1 April 2020 (2020-04-01), & EXPERIMENTAL BIOLOGY MEETING; SAN DIEGO, CA, USA; APRIL 21 -25, 2018, pages 5435 - 5452, XP055837326, ISSN: 0892-6638, Retrieved from the Internet <URL:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7136136/pdf/FSB2-34-5435.pdf> DOI: 10.1096/fj.201902610R *
MULLER PAJVOUSDEN KH: "p53 mutations in cancer", NAT CELL BIO, vol. 15, 2013, pages 2 - 8
OLINER J.D.: "Cancer. p53, guardian of the genome", NATURE, vol. 358, no. 6381, 1992, pages 15 - 16
PARKS ET AL., BIOORG. MED. CHEM. LETT., vol. 15, 2005, pages 765 - 770
PETITJEAN AMATHE EKATO SISHIOKA CTAVTIGIAN SVHAINAUT POLIVIER M: "Impact of mutant P53 functional properties on TP53 mutation patterns and tumor phenotype: lessons from recent developments in the IARC TP53 database", HUM MUTAT, vol. 28, no. 6, June 2007 (2007-06-01), pages 622 - 9
RIVLIN NKOIFMAN GROTTER V: "p53 orchestrates between normal differentiation and cancer", SEMIN CANCER BIOL, vol. 32, 2015, pages 10 - 17, XP029129005, DOI: 10.1016/j.semcancer.2013.12.006
SHANGARY ET AL., ANN. REV. PHARMACOL. TOXICOL., vol. 49, 2008, pages 223 - 241
SHANGARY ET AL., MOL. CANCER THER., vol. 7, 2008, pages 1533 - 1542
SHANGARY ET AL., PROC. NATL. ACAD. SCI. USA, vol. 105, 2008, pages 3933 - 3938
STOLL ET AL., BIOCHEMISTRY, vol. 40, 2001, pages 336 - 44
TEFFERI A ET AL.: "CALR and ASXL1 mutations-based molecular prognostication in primary myelofibrosis: an international study of 570 patients", LEUKEMIA, vol. 28, no. 7, 2014, pages 1494 - 1500
TEFFERI A ET AL.: "Targeted deep sequencing in polycythemia vera and essential thrombocythemia", BLOODADV, vol. 1, no. 1, 2016, pages 21 - 30
VANNUCCHI AM ET AL.: "Mutations and prognosis in primary myelofibrosis", LEUKEMIA, vol. 27, no. 9, 2013, pages 1861 - 1869, XP037785377, DOI: 10.1038/leu.2013.119
VANNUCCHI ET AL., CA CANCER J. CLIN., vol. 59, 2009, pages 171 - 191
VASSILEV LT ET AL.: "In vivo activation of the p53 pathway by small-molecule antagonists of MDM2", SCIENCE, vol. 303, no. 5659, 2004, pages 844 - 848, XP002338500, DOI: 10.1126/science.1092472
YIN ET AL., ANGEW. CHEM. INT. ED. ENGL., vol. 44, 2005, pages 2704 - 2707

Similar Documents

Publication Publication Date Title
US10704108B2 (en) Genetic alterations in isocitrate dehydrogenase and other genes in malignant glioma
Tyner et al. Functional genomic landscape of acute myeloid leukaemia
US20220243283A1 (en) Methods for typing of lung cancer
EP3149209B1 (en) Methods for typing of lung cancer
JP2009544583A (en) Cancer treatment method using TAK1 inhibitor
US20240033265A2 (en) Treatments for mutations in acute myeloid leukemia
KR20190039730A (en) Use of mitochondrial activity inhibitors for the treatment of acute myeloid leukemia with poor prognosis
WO2022219128A1 (en) In vitro tests allowing to identify the potential for mdm2 inhibitors to induce the selection of mutations in patients suffering from a myeloproliferative neoplasm
WO2017079695A9 (en) Gene expression patterns to predict responsiveness to virotherapy in cancer indications
AU2015202486A1 (en) Genetic alterations in isocitrate dehydrogenase and other genes in malignant glioma
EP4136262A1 (en) Prognosis method of acute myeloid leukaemia
WO2023244582A1 (en) Lung cancer-related biomarkers and methods of using the same
EP3665307A1 (en) Materials and methods for stratifying and treating cancers
Selvarajah et al. Gene expression profiling and pathways associated with high metastatic potential in canine osteosarcoma
AU2013201554A1 (en) Genetic alterations in isocitrate dehydrogenase and other genes in malignant glioma

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22722828

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22722828

Country of ref document: EP

Kind code of ref document: A1