WO2020115261A1 - Méthodes et compositions pour le traitement du mélanome - Google Patents

Méthodes et compositions pour le traitement du mélanome Download PDF

Info

Publication number
WO2020115261A1
WO2020115261A1 PCT/EP2019/083926 EP2019083926W WO2020115261A1 WO 2020115261 A1 WO2020115261 A1 WO 2020115261A1 EP 2019083926 W EP2019083926 W EP 2019083926W WO 2020115261 A1 WO2020115261 A1 WO 2020115261A1
Authority
WO
WIPO (PCT)
Prior art keywords
melanoma
lyn
resistant
subject
treatment
Prior art date
Application number
PCT/EP2019/083926
Other languages
English (en)
Inventor
Robert Ballotti
Corine Bertolotto
Céline GAUDEL
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
Université Cote D'azur
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), Université Cote D'azur filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Publication of WO2020115261A1 publication Critical patent/WO2020115261A1/fr

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • G01N33/5743Specifically defined cancers of skin, e.g. melanoma
    • 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
    • C12Q2600/158Expression markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/91Transferases (2.)
    • G01N2333/912Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • G01N2333/91205Phosphotransferases in general
    • G01N2333/9121Phosphotransferases in general with an alcohol group as acceptor (2.7.1), e.g. general tyrosine, serine or threonine kinases
    • G01N2333/91215Phosphotransferases in general with an alcohol group as acceptor (2.7.1), e.g. general tyrosine, serine or threonine kinases with a definite EC number (2.7.1.-)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/10Screening for compounds of potential therapeutic value involving cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/56Staging of a disease; Further complications associated with the disease

Definitions

  • the invention is in the field of oncology, more particularly the invention relates to methods and compositions for treating melanoma.
  • Cutaneous melanoma represents 3% of all cancers, with 8,000 new cases in France and 12,000 new cases in the United Kingdom in 2010. Melanoma has a favorable prognosis when it is diagnosed early, but at the metastatic stage, the prognosis is still very pejorative.
  • BRAF inhibitors Zelboraf, Tafmlar
  • MEK inhibitor Tafmlar plus Mekinist
  • the kinases of the SRC family comprise 8 members and have long been known to be oncogenes [4] Recent reports showed that resistance to BRAF inhibitors can be bypassed by a combination of BRAF inhibitor and SRC inhibitors [5-7] With the aims of deciphering the molecular mechanisms involved in the implementation of resistances and to identify pharmacologically targetable players, the inventors compared the expression of kinases of the SRC family members in melanoma cells isolated from patients before treatment with targeted therapy and after acquisition of resistance to BRAF inhibitors.
  • the invention relates to methods for treating melanoma and therapy-resistant melanoma in a subject in need thereof comprising the step of administering said subject with a therapeutically effective amount of an activator of LYN.
  • the present invention is defined by the claims.
  • inventors have shown that the loss of expression of LYN is correlated with BRAF inhibitors resistance. Secondly, inventors have shown that overexpression of LYN in resistant cells that have lost LYN causes inhibition of their proliferation and migratory capacity.
  • the invention relates to a method for predicting whether a subject suffering from a melanoma is or is at risk of having resistant melanoma comprising the steps of i) quantifying the expression level of LYN in a biological sample obtained from the subject; ii) comparing the expression level quantified at step i) with a predetermined reference value and iii) concluding that the subject has or is at risk of suffering from resistant melanoma when the level determined at step i) is lower than the predetermined reference value.
  • melanoma also known as malignant melanoma
  • malignant melanoma refers to a type of cancer that develops from the pigment-containing cells, called melanocytes.
  • melanoma There are three general categories of melanoma: 1) cutaneous melanoma which corresponds to melanoma of the skin; it is the most common type of melanoma; 2) mucosal melanoma which can occur in any mucous membrane of the body, including the nasal passages, the throat, the vagina, the anus, or in the mouth; and 3) ocular melanoma also known as uveal melanoma or choroidal melanoma, is a rare form of melanoma that occurs in the eye.
  • the melanoma is cutaneous melanoma.
  • the term“subject” denotes a mammal, such as a rodent, a feline, a canine, and a primate.
  • the subject according to the invention is a human. More particularly, the subject according to the invention has or is susceptible to have melanoma. In particular embodiment, the subject has or is susceptible to have cutaneous melanoma. In a particular embodiment, the subject has or is susceptible to have metastatic melanoma.
  • the subject has or is susceptible to have resistant melanoma.
  • resistant melanoma refers to melanoma which does not respond to a treatment.
  • the cancer may be resistant at the beginning of treatment or it may become resistant during treatment.
  • the resistance to drug leads to rapid progression of metastatic of melanoma.
  • resistant melanoma cell refers to cell which does not respond to a treatment.
  • the term“sensitive melanoma cell” refers to cell which does respond to a treatment.
  • the melanoma is resistant to BRAF inhibitors.
  • BRAF is a member of the Raf kinase family of serine/threonine-specific protein kinases. This protein plays a role in regulating the MAP kinase / ERKs signaling pathway, which affects cell division, differentiation, and secretion.
  • a number of mutations in BRAF are known. In particular, the V600E mutation is prominent.
  • mutations which have been found are R461I, I462S, G463E, G463V, G465A, G465E, G465V, G468A, G468E, N580S, E585K, D593V, F594L, G595R, L596V, T598I, V599D, V599E, V599K, V599R, K600E, A727V, and most of these mutations are clustered to two regions: the glycine-rich P loop of the N lobe and the activation segment and flanking regions.
  • the BRAF mutation is V600E.
  • the melanoma is resistant to MEK inhibitors.
  • MEK refers to Mitogen-activated protein kinase kinase, also known as MAP2K, MEK, MAPKK. It is a kinase enzyme which phosphorylates mitogen-activated protein kinase (MAPK). MEK is activated in melanoma.
  • the melanoma is resistant to NRAS inhibitors.
  • the NRAS gene is in the Ras family of oncogene and involved in regulating cell division. NRAS mutations in codons 12, 13, and 61 arise in 15-20 % of all melanomas.
  • the melanoma is resistant to immune checkpoint inhibitors.
  • immune checkpoint inhibitor refers to molecules that totally or partially reduce, inhibit, interfere with or modulate one or more immune checkpoint proteins.
  • immuno checkpoint protein has its general meaning in the art and refers to a molecule that is expressed by T cells in that either turn up a signal (stimulatory checkpoint molecules) or turn down a signal (inhibitory checkpoint molecules). Immune checkpoint molecules are recognized in the art to constitute immune checkpoint pathways similar to the CTLA-4 and PD-1 dependent pathways (see e.g. Pardoll, 2012. Nature Rev Cancer 12:252-264; Mellman et al. 2011. Nature 480:480- 489).
  • stimulatory checkpoint examples include CD27 CD28 CD40, CD122, CD137, 0X40, GITR, and ICOS.
  • inhibitory checkpoint molecules examples include A2AR, B7-H3, B7-H4, BTLA, CTLA-4, CD277, IDO, KIR, PD-1, LAG-3, TIM-3 and VISTA.
  • the Adenosine A2A receptor (A2AR) is regarded as an important checkpoint in cancer therapy because adenosine in the immune microenvironment, leading to the activation of the A2a receptor, is negative immune feedback loop and the tumor microenvironment has relatively high concentrations of adenosine.
  • B7-H3 also called CD276, was originally understood to be a co-stimulatory molecule but is now regarded as co-inhibitory.
  • B7-H4 also called VTCN1
  • B and T Lymphocyte Attenuator (BTLA) and also called CD272 has HVEM (Herpesvirus Entry Mediator) as its ligand.
  • HVEM Herpesvirus Entry Mediator
  • Surface expression of BTLA is gradually downregulated during differentiation of human CD8+ T cells from the naive to effector cell phenotype, however tumor-specific human CD8+ T cells express high levels of BTLA.
  • CTLA-4 Cytotoxic T -Lymphocyte- Associated protein 4 and also called CD152.
  • IDO Indoleamine 2,3-dioxygenase
  • TDO tryptophan catabolic enzyme
  • TDO tryptophan 2,3 -dioxygenase
  • KIR Killer-cell Immunoglobulin-like Receptor
  • LAG3, Lymphocyte Activation Gene-3 works to suppress an immune response by action to Tregs as well as direct effects on CD8+ T cells.
  • PD- 1 Programmed Death 1 (PD-1) receptor
  • PD-L1 and PD-L2 This checkpoint is the target of Merck & Co.'s melanoma drug Keytruda, which gained FDA approval in September 2014.
  • An advantage of targeting PD-1 is that it can restore immune function in the tumor microenvironment.
  • TIM-3 short for T-cell Immunoglobulin domain and Mucin domain 3, expresses on activated human CD4+ T cells and regulates Thl and Thl7 cytokines.
  • TIM-3 acts as a negative regulator of Thl/Tcl function by triggering cell death upon interaction with its ligand, galectin-9.
  • VISTA Short for V-domain Ig suppressor of T cell activation, VISTA is primarily expressed on hematopoietic cells so that consistent expression of VISTA on leukocytes within tumors may allow VISTA blockade to be effective across a broad range of solid tumors. Tumor cells often take advantage of these checkpoints to escape detection by the immune system. Thus, inhibiting a checkpoint protein on the immune system may enhance the anti-tumor T-cell response.
  • an immune checkpoint inhibitor refers to any compound inhibiting the function of an immune checkpoint protein. Inhibition includes reduction of function and full blockade.
  • the immune checkpoint inhibitor could be an antibody, synthetic or native sequence peptides, small molecules or aptamers which bind to the immune checkpoint proteins and their ligands.
  • the immune checkpoint inhibitor is an antibody.
  • antibodies are directed against A2AR, B7-H3, B7-H4, BTLA, CTLA-4, CD277, IDO, KIR, PD-1, LAG-3, TIM-3 or VISTA.
  • the immune checkpoint inhibitor is an anti-PD-1 antibody such as described in WO2011082400, W02006121168, W02015035606, W02004056875, W02010036959, W02009114335, W02010089411, WO2008156712, WO2011110621, WO2014055648 and WO2014194302.
  • anti-PD-1 antibodies which are commercialized: Nivolumab (Opdivo®, BMS), Pembrolizumab (also called Lambrolizumab, KEYTRUDA® or MK-3475, MERCK).
  • the immune checkpoint inhibitor is an anti-PD-Ll antibody such as described in WO2013079174, W02010077634, W02004004771, WO2014195852, W02010036959, WO2011066389, W02007005874, W02015048520, US8617546 and WO2014055897.
  • anti-PD-Ll antibodies which are on clinical trial: Atezolizumab (MPDL3280A, Genentech/Roche), Durvalumab (AZD9291, AstraZeneca), Avelumab (also known as MSB0010718C, Merck) and BMS-936559 (BMS).
  • the immune checkpoint inhibitor is an anti-PD-L2 antibody such as described in US7709214, US7432059 and US8552154.
  • the immune checkpoint inhibitor inhibits Tim-3 or its ligand.
  • the immune checkpoint inhibitor is an anti-Tim-3 antibody such as described in WO03063792, WO2011155607, WO2015117002, WO2010117057 and W02013006490.
  • the immune checkpoint inhibitor is a small organic molecule.
  • small organic molecule refers to a molecule of a size comparable to those organic molecules generally used in pharmaceuticals.
  • small organic molecules range in size up to about 5000 Da, more preferably up to 2000 Da, and most preferably up to about 1000 Da.
  • the small organic molecules interfere with transduction pathway of A2AR, B7-H3, B7-H4, BTLA, CTLA-4, CD277, IDO, KIR, PD-1, LAG-3, TIM-3 or VISTA.
  • small organic molecules interfere with transduction pathway of PD-1 and Tim-3.
  • they can interfere with molecules, receptors or enzymes involved in PD-1 and Tim-3 pathway.
  • the small organic molecules interfere with Indoleamine- pyrrole 2,3-dioxygenase (IDO) inhibitor.
  • IDO is involved in the tryptophan catabolism (Liu et al 2010, Vacchelli et al 2014, Zhai et al 2015). Examples of IDO inhibitors are described in WO 2014150677.
  • IDO inhibitors include without limitation 1 -methyl-tryptophan (IMT), b- (3-benzofuranyl)-alanine, P-(3-benzo(b)thienyl)-alanine), 6-nitro-tryptophan, 6- fluoro-tryptophan, 4-methyl-tryptophan, 5 -methyl tryptophan, 6-methyl-tryptophan, 5- m ethoxy-tryptophan, 5 -hydroxy-tryptophan, indole 3-carbinol, 3,3'- diindolylmethane, epigallocatechin gallate, 5-Br-4-Cl-indoxyl 1,3-diacetate, 9- vinylcarbazole, acemetacin, 5- bromo-tryptophan, 5-bromoindoxyl diacetate, 3- Amino-naphtoic acid, pyrrolidine dithiocarbamate, 4-phenylimidazole a brassinin derivative, a thioh
  • the IDO inhibitor is selected from 1 -methyl-tryptophan, b-(3- benzofuranyl)-alanine, 6-nitro-L-tryptophan, 3- Amino-naphtoic acid and b-[3- benzo(b)thienyl] -alanine or a derivative or prodrug thereof.
  • the inhibitor of IDO is Epacadostat, (INCB24360, INCB024360) has the following chemical formula in the art and refers to -N-(3-bromo-4- fluorophenyl)-N'-hydroxy-4- ⁇ [2-(sulfamoylamino)-ethyl]amino ⁇ -l,2,5-oxadiazole-3 carboximidamide :
  • the inhibitor is BGB324, also called R428, such as described in W02009054864, refers to lH-1, 2, 4-Triazole-3, 5-diamine, l-(6,7-dihydro-5H- benzo[6,7]cyclohepta[l,2-c]pyridazin-3-yl)-N3-[(7S)-6,7,8,9-tetrahydro-7-(l-pyrrolidinyl)- 5H-benzocyclohepten-2-yl]- and has the following formula in the art:
  • the inhibitor is CA-170 (or AUPM-170): an oral, small molecule immune checkpoint antagonist targeting programmed death ligand-1 (PD-L1) and V- domain Ig suppressor of T cell activation (VISTA) (Liu et al 2015).
  • PD-170 or AUPM-170
  • VISTA V- domain Ig suppressor of T cell activation
  • the immune checkpoint inhibitor is an aptamer.
  • the aptamers are directed against A2AR, B7-H3, B7-H4, BTLA, CTLA-4, CD277, IDO, KIR, PD-1, LAG-3, TIM-3 or VISTA.
  • aptamers are DNA aptamers such as described in Prodeus et al 2015.
  • a major disadvantage of aptamers as therapeutic entities is their poor pharmacokinetic profiles, as these short DNA strands are rapidly removed from circulation due to renal filtration.
  • aptamers according to the invention are conjugated to with high molecular weight polymers such as polyethylene glycol (PEG).
  • PEG polyethylene glycol
  • the aptamer is an anti-PD-1 aptamer.
  • the anti-PD-1 aptamer is MP7 pegylated as described in Prodeus et al 2015.
  • the term "predicting the risk” refers to assessing the probability according to which the subject as referred to herein will develop resistant melanoma. As will be understood by those skilled in the art, such an assessment is usually not intended to be correct for 100% of the subjects to be investigated.
  • the term“SRC kinase family” refers to family of non-receptor tyrosine kinases implicate in cancer and immune system dysfunction.
  • SRC kinase family include the following nine kinases in mammals: Src, Yes, Fyn, and Fgr, forming the SrcA subfamily, Lck, Hck, Blk, and Lyn in the SrcB subfamily, and Frk in its own subfamily. These are non-receptor protein kinases that range in molecular mass from 52 to 62 kD.
  • the SRC kinase is LYN.
  • LYN refers to Tyrosine-protein kinase Lyn, a protein that is encoded in human by the LYN gene. Lyn is mainly expressed in hematopoietic cells, in neural tissues, liver and adipose tissue.
  • the naturally occurring human LYN gene has a nucleotide sequence as shown in Genbank Accession number NM_002350.3 and the naturally occurring human LYN protein has an aminoacid sequence as shown in Genbank Accession numbers NP_002341.1 for LYN isoform A and NP_001104567.1 for LYN isoform B.
  • the term“expression level” refers to the expression level of LYN.
  • the expression level of the LYN gene may be determined by any technology known by a person skilled in the art.
  • each gene expression level may be measured at the genomic and/or nucleic and/or protein level.
  • the expression level of gene is determined by measuring the amount of nucleic acid transcripts of each gene.
  • the expression level is determined by measuring the amount of each gene corresponding protein. The amount of nucleic acid transcripts can be measured by any technology known by a man skilled in the art.
  • the measure may be carried out directly on an extracted messenger RNA (mRNA) sample, or on retrotranscribed complementary DNA (cDNA) prepared from extracted mRNA by technologies well-known in the art.
  • mRNA messenger RNA
  • cDNA retrotranscribed complementary DNA
  • the amount of nucleic acid transcripts may be measured using any technology known by a man skilled in the art, including nucleic microarrays, quantitative PCR, microfluidic cards, and hybridization with a labelled probe.
  • the expression level is determined by using quantitative PCR. Quantitative, or real-time, PCR is a well-known and easily available technology for those skilled in the art and does not need a precise description. Methods for determining the quantity of mRNA are well known in the art.
  • the nucleic acid contained in the biological sample is first extracted according to standard methods, for example using lytic enzymes or chemical solutions or extracted by nucleic-acid-binding resins following the manufacturer's instructions.
  • the extracted mRNA is then detected by hybridization (e. g., Northern blot analysis) and/or amplification (e.g., RT-PCR).
  • hybridization e. g., Northern blot analysis
  • amplification e.g., RT-PCR
  • RT-PCR e.g., RT-PCR
  • RT-PCR e.g., RT-PCR
  • RT-PCR e.g., RT-PCR
  • RT-PCR e.g., RT-PCR
  • RT-PCR e.g., RT-PCR
  • RT-PCR e.g., RT-PCR
  • RT-PCR e.g., RT-PCR
  • RT-PCR e.g., RT-PCR
  • Nucleic acids having at least 10 nucleotides and exhibiting sequence complementarity or homology to the mRNA of interest herein find utility as hybridization probes or amplification primers. It is understood that such nucleic acids do not need to be identical, but are typically at least about 80% identical to the homologous region of comparable size, more preferably 85% identical and even more preferably 90-95% identical. In certain embodiments, it will be advantageous to use nucleic acids in combination with appropriate means, such as a detectable label, for detecting hybridization. A wide variety of appropriate indicators are known in the art including, fluorescent, radioactive, enzymatic or other ligands (e. g. avidin/biotin).
  • Probes typically comprise single-stranded nucleic acids of between 10 to 1000 nucleotides in length, for instance of between 10 and 800, more preferably of between 15 and 700, typically of between 20 and 500.
  • Primers typically are shorter single-stranded nucleic acids, of between 10 to 25 nucleotides in length, designed to perfectly or almost perfectly match a nucleic acid of interest, to be amplified.
  • the probes and primers are“specific” to the nucleic acids they hybridize to, i.e. they preferably hybridize under high stringency hybridization conditions (corresponding to the highest melting temperature Tm, e.g., 50 % formamide, 5x or 6x SCC.
  • SCC is a 0.15 M NaCl, 0.015 M Na-citrate).
  • the nucleic acid primers or probes used in the above amplification and detection method may be assembled as a kit.
  • a kit includes consensus primers and molecular probes.
  • a kit also includes the components necessary to determine if amplification has occurred.
  • the kit may also include, for example, PCR buffers and enzymes; positive control sequences, reaction control primers; and instructions for amplifying and detecting the specific sequences.
  • the method of the invention comprises the steps of providing total RNAs extracted from a biological sample and subjecting the RNAs to amplification and hybridization to specific probes, more particularly by means of a quantitative or semi-quantitative RT-PCR.
  • the expression level is determined by DNA chip analysis.
  • Such DNA chip or nucleic acid microarray consists of different nucleic acid probes that are chemically attached to a substrate, which can be a microchip, a glass slide or a microsphere-sized bead.
  • a microchip may be constituted of polymers, plastics, resins, polysaccharides, silica or silica-based materials, carbon, metals, inorganic glasses, or nitrocellulose.
  • Probes comprise nucleic acids such as cDNAs or oligonucleotides that may be about 10 to about 60 base pairs.
  • a biological sample from a test subject optionally first subjected to a reverse transcription, is labelled and contacted with the microarray in hybridization conditions, leading to the formation of complexes between target nucleic acids that are complementary to probe sequences attached to the microarray surface.
  • the labelled hybridized complexes are then detected and can be quantified or semi- quantified. Labelling may be achieved by various methods, e.g. by using radioactive or fluorescent labelling.
  • Many variants of the microarray hybridization technology are available to the man skilled in the art (see e.g. the review by Hoheisel, Nature Reviews, Genetics, 2006, 7:200-210).
  • biological sample refers to any sample obtained from a subject, such as a serum sample, a plasma sample, a urine sample, a blood sample, a lymph sample, or a tissue biopsy.
  • biological sample for the determination of an expression level include samples such as a blood sample, a lymph sample, or a biopsy.
  • the biological sample is a blood sample, more particularly, circulating tumor cells (CTCs). Detection of CTCs are available to the man skilled in the art (see e.g. the article by Long et al, Cancer Medecine, 2016, 5(6): 1022-1030 [9]).
  • the predetermined reference value is a threshold value or a cut-off value.
  • a “threshold value” or “cut-off value” can be determined experimentally, empirically, or theoretically.
  • a threshold value can also be arbitrarily selected based upon the existing experimental and/or clinical conditions, as would be recognized by a person of ordinary skilled in the art. For example, retrospective measurement of cell densities in properly banked historical subject samples may be used in establishing the predetermined reference value. The threshold value has to be determined in order to obtain the optimal sensitivity and specificity according to the function of the test and the benefit/risk balance (clinical consequences of false positive and false negative).
  • the optimal sensitivity and specificity can be determined using a Receiver Operating Characteristic (ROC) curve based on experimental data.
  • ROC Receiver Operating Characteristic
  • the full name of ROC curve is receiver operator characteristic curve, which is also known as receiver operation characteristic curve. It is mainly used for clinical biochemical diagnostic tests.
  • ROC curve is a comprehensive indicator that reflects the continuous variables of true positive rate (sensitivity) and false positive rate (1 -specificity). It reveals the relationship between sensitivity and specificity with the image composition method.
  • a series of different cut-off values are set as continuous variables to calculate a series of sensitivity and specificity values. Then sensitivity is used as the vertical coordinate and specificity is used as the horizontal coordinate to draw a curve. The higher the area under the curve (AUC), the higher the accuracy of diagnosis.
  • AUC area under the curve
  • the point closest to the far upper left of the coordinate diagram is a critical point having both high sensitivity and high specificity values.
  • the AUC value of the ROC curve is between 1.0 and 0.5. When AUC>0.5, the diagnostic result gets better and better as AUC approaches 1. When AUC is between 0.5 and 0.7, the accuracy is low. When AUC is between 0.7 and 0.9, the accuracy is moderate.
  • This algorithmic method is preferably done with a computer.
  • Existing software or systems in the art may be used for the drawing of the ROC curve, such as: MedCalc 9.2.0.1 medical statistical software, SPSS 9.0, ROCPOWER.SAS, DESIGNROC.FOR, MULTIREADER POWER S AS, CREATE-ROC.SAS, GB STAT VIO.O (Dynamic Microsystems, Inc. Silver Spring, Md., USA), etc.
  • the predetermined reference value is determined by carrying out a method comprising the steps of
  • step c) classifying said tumor tissue samples in two groups for one specific arbitrary quantification value provided at step c), respectively: (i) a first group comprising tumor tissue samples that exhibit a quantification value for level that is lower than the said arbitrary quantification value contained in the said serial of quantification values; (ii) a second group comprising tumor tissue samples that exhibit a quantification value for said level that is higher than the said arbitrary quantification value contained in the said serial of quantification values; whereby two groups of tumor tissue samples are obtained for the said specific quantification value, wherein the tumor tissue samples of each group are separately enumerated;
  • step g) reiterating steps f) and g) until every arbitrary quantification value provided at step d) is tested; h) setting the said predetermined reference value as consisting of the arbitrary quantification value for which the highest statistical significance (most significant P-value obtained with a log-rank test, significance when P ⁇ 0.05) has been calculated at step g).
  • the cell density has been assessed for 100 tumor tissue samples of 100 subjects.
  • the 100 samples are ranked according to the cell density.
  • Sample 1 has the highest density and sample 100 has the lowest density.
  • a first grouping provides two subsets: on one side sample Nr 1 and on the other side the 99 other samples.
  • the next grouping provides on one side samples 1 and 2 and on the other side the 98 remaining samples etc., until the last grouping: on one side samples 1 to 99 and on the other side sample Nr 100.
  • Kaplan-Meier curves are prepared for each of the 99 groups of two subsets. Also for each of the 99 groups, the p value between both subsets was calculated (log-rank test).
  • the predetermined reference value is then selected such as the discrimination based on the criterion of the minimum P-value is the strongest.
  • the cell density corresponding to the boundary between both subsets for which the P-value is minimum is considered as the predetermined reference value.
  • the predetermined reference value is not necessarily the median value of cell densities.
  • the predetermined reference value thus allows discrimination between a poor and a good prognosis with respect to DFS and OS for a subject. Practically, high statistical significance values (e.g. low P values) are generally obtained for a range of successive arbitrary quantification values, and not only for a single arbitrary quantification value.
  • a range of values is provided instead of using a definite predetermined reference value. Therefore, a minimal statistical significance value (minimal threshold of significance, e.g. maximal threshold P value) is arbitrarily set and a range of a plurality of arbitrary quantification values for which the statistical significance value calculated at step g) is higher (more significant, e.g. lower P-value) are retained, so that a range of quantification values is provided.
  • This range of quantification values includes a "cut-off value as described above. For example, according to this specific embodiment of a "cut-off value, the outcome can be determined by comparing the cell density with the range of values which are identified.
  • a cut-off value thus consists of a range of quantification values, e.g. centered on the quantification value for which the highest statistical significance value is found (e.g. generally the minimum P-value which is found).
  • the invention relates to a method for treating melanoma in a subject in need thereof comprising a step of administering said subject with a therapeutically effective amount of an activator of LYN.
  • the subject is identified as having melanoma resistance by performing the method as described above.
  • the invention relates to a method for treating resistant melanoma in a subject in need thereof comprising a step of administering said subject with a therapeutically effective amount of an activator of LYN.
  • the subject is identified as having melanoma resistant to a classical treatment.
  • the subject is identified as having BRAF inhibitors resistance.
  • the subject is identified as having MEK inhibitors resistance.
  • the subject is identified as having NRAS inhibitors resistance.
  • the subject is identified as having immune checkpoint inhibitors resistance.
  • the activators of LYN may be combined with an immune checkpoint inhibitor.
  • the present invention relates to a method for treating melanoma and/or resistant melanoma in a subject in need thereof comprising a step of administering said subject with a therapeutically effective amount of an activator of LYN and of an immune checkpoint inhibitor.
  • the terms“treating” or“treatment” refer to both prophylactic or preventive treatment as well as curative or disease modifying treatment, including treatment of subject at risk of contracting the disease or suspected to have contracted the disease as well as subject 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., pain, disease manifestation, etc.]).
  • the term“activator of LYN” refers to a natural or synthetic compound that has a biological effect to increase the activity of LYN. More particularly, such compound induces phosphorylation of Tyr396 in the activation loop of LYN that generates a highly active LYNand/or increases the kinase activity of LYN toward its natural (Cortactin, HS1) or synthetic substrates (Poly (Glu4-Tyr) or SRCtide (GAELPPEF A AQLRKIGDK V Y C (SEQ ID NO: 1))
  • the activator of LYN is a LYN allosteric activator. In a particular embodiment, the activator of LYN is tolimidone.
  • tolimidone also known as MLR-1023 or 2(1H)- pyrimidinone,5-(3-methylphenoxy) has the formula Cl 1H10N2O2 and the following structure in the art:
  • the activator of LYN is a derivative of MLR-1023 as describes in W02010/003055.
  • the activator of LYN is a peptide, petptidomimetic, small organic molecule, antibody, aptamers, siRNA or antisense oligonucleotide.
  • peptido mimetic refers to a small protein-like chain designed to mimic a peptide.
  • the activator of LYN is an aptamer.
  • Aptamers are a class of molecule that represents an alternative to antibodies in term of molecular recognition. Aptamers are oligonucleotide or oligopeptide sequences with the capacity to recognize virtually any class of target molecules with high affinity and specificity.
  • the activator of LYN is a small organic molecule.
  • small organic molecule refers to a molecule of a size comparable to those organic molecules generally used in pharmaceuticals. The term excludes biological macromolecules (e.g., proteins, nucleic acids, etc.). Preferred small organic molecules range in size up to about 5000 Da, more preferably up to 2000 Da, and most preferably up to about 1000 Da.
  • the activator of LYN is an antibody.
  • antibody is used in the broadest sense and specifically covers monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g. bispecific antibodies) formed from at least two intact antibodies, and antibody fragments so long as they exhibit the desired biological activity.
  • the term includes antibody fragments that comprise an antigen binding domain such as Fab', Fab, F(ab')2, single domain antibodies (DABs), TandAbs dimer, Fv, scFv (single chain Fv), dsFv, ds-scFv, Fd, linear antibodies, minibodies, diabodies, bispecific antibody fragments, bibody, tribody (scFv-Fab fusions, bispecific or trispecific, respectively); sc-diabody; kappa(lamda) bodies (scFv-CL fusions); BiTE (Bispecific T-cell Engager, scFv-scFv tandems to attract T cells); DVD-Ig (dual variable domain antibody, bispecific format); SIP (small immunoprotein, a kind of minibody); SMIP ("small modular immunopharmaceutical” scFv-Fc dimer; DART (ds-stabilized diabody "Dual Affinity ReTargeting"
  • Antibodies can be fragmented using conventional techniques. For example, F(ab')2 fragments can be generated by treating the antibody with pepsin. The resulting F(ab')2 fragment can be treated to reduce disulfide bridges to produce Fab' fragments. Papain digestion can lead to the formation of Fab fragments.
  • Fab, Fab' and F(ab')2, scFv, Fv, dsFv, Fd, dAbs, TandAbs, ds-scFv, dimers, minibodies, diabodies, bispecific antibody fragments and other fragments can also be synthesized by recombinant techniques or can be chemically synthesized. Techniques for producing antibody fragments are well known and described in the art. For example, each of Beckman et al, 2006; Holliger & Hudson, 2005; Le Gall et al, 2004; Reff & Heard, 2001 ; Reiter et al., 1996; and Young et al., 1995 further describe and enable the production of effective antibody fragments.
  • the antibody is a“chimeric” antibody as described in U.S. Pat. No. 4,816,567.
  • the antibody is a humanized antibody, such as described U.S. Pat. Nos. 6,982,321 and 7,087,409.
  • the antibody is a human antibody.
  • A“human antibody” such as described in US 6,075, 181 and 6, 150,584.
  • the antibody is a single domain antibody such as described in EP 0 368 684, WO 06/030220 and WO 06/003388.
  • the activator of LYN is a monoclonal antibody.
  • Monoclonal antibodies can be prepared and isolated using any technique that provides for the production of antibody molecules by continuous cell lines in culture. Techniques for production and isolation include but are not limited to the hybridoma technique, the human B-cell hybridoma technique and the EBV-hybridoma technique.
  • the activator is an intrabody having specificity for LYN.
  • the term "intrabody” generally refer to an intracellular antibody or antibody fragment.
  • Antibodies in particular single chain variable antibody fragments (scFv), can be modified for intracellular localization. Such modification may entail for example, the fusion to a stable intracellular protein, such as, e.g., maltose binding protein, or the addition of intracellular trafficking/localization peptide sequences, such as, e.g., the endoplasmic reticulum retention.
  • the intrabody is a single domain antibody.
  • the antibody according to the invention is a single domain antibody.
  • sdAb single domain antibody
  • VHH single domain antibody
  • sdAb single domain antibody
  • VHH single domain antibody
  • LYN activator In order to test the functionality of a putative LYN activator a test is necessary. For that purpose, to identify LYN activator an in vitro kinase assay will be used. In brief, recombinant LYN kinase is incubated with a synthetic substrate (SRCtides), ATP as phosphate donor, and increasing concentration of the potential activators. MLR- 1023 is used as reference agent, and Staurosporine, (a known nonspecific LYN inhibitor) as inhibition control.
  • SRCtides synthetic substrate
  • MLR- 1023 is used as reference agent
  • Staurosporine (a known nonspecific LYN inhibitor) as inhibition control.
  • melanoma treatment with pharmacological LYN activator can overcome resistance to drugs targeting oncogenic BRAF.
  • the resistance of cancer for the medication is caused by mutations in the genes which are involved in the proliferation, divisions or differentiation of cells or by phenotypic switch with a transcriptional profile favoring the resistance to treatment.
  • the resistance of melanoma is caused by the mutations (single or double) in the following genes: BRAF, MEK or NRAS.
  • the resistance can be also caused by a double-negative BRAF and NRAS mutation or phenotypic switch.
  • the melanoma is resistant to a treatment with the inhibitors of BRAF mutations.
  • the melanoma is resistant to a treatment.
  • the melanoma is resistant to a treatment with the inhibitors of BRAF.
  • the melanoma is resistant to a treatment with dabrafenib also known as tafmlar which is commercialized by Novartis.
  • the melanoma is resistant to a treatment with vemurafenib.
  • Vemurafenib also known as PLX4032, RG7204 ou R05185426 and commercialized by Roche as Zelboraf.
  • the melanoma is resistant to a treatment with dacarbazine.
  • dacarbazine also known as imidazole carboxamide is commercialized as DTIC-Dome by Bayer.
  • the melanoma is resistant to a treatment with the inhibitors of MEK.
  • the inhibitors of MEK are well known in the art.
  • the melanoma is resistant to a treatment with trametinib also known as mekinist which is commercialized by GSK.
  • the melanoma is resistant to a treatment with cobimetinib also known as cotellic commercialized by Genentech.
  • the melanoma is resistant to a treatment with Binimetinib also knowns as MEK162, ARRY-162 is developed by Array Biopharma.
  • the melanoma is resistant to a treatment with the inhibitors of NRAS. In a particular embodiment, the melanoma is resistant to a treatment with salirasib commercialized by Concordia Pharmaceuticals.
  • the inhibitors of BRAF mutation or MEK are used to treat the melanoma with NRAS mutations. In a particular embodiment, the melanoma is resistant in which double-negative BRAF and NRAS mutant melanoma.
  • the melanoma is resistant to a combined treatment.
  • the terms“combined treatment”,“combined therapy” or“therapy combination” refer to a treatment that uses more than one medication.
  • the combined therapy may be dual therapy or bi-therapy.
  • the melanoma is resistant to a combined treatment characterized by using an inhibitor of BRAF mutation and an inhibitor of MEK or an inhibitor of BRAF mutation and an inhibitor of NRAS as described above.
  • the combined treatment may be a combination of Vemurafenib (BRAFi) and Cobimetinib (MEKi), or a combination of Dabrafenib (BRAFi) and Trametinib (MEKi), or a combination of Cobimetinib Encorafenib (BRAFi) and Binimetinib(MEKi).
  • the melanoma is resistant to a treatment with an immune checkpoint inhibitor.
  • immune checkpoint inhibitor as used herein, is defined above.
  • the melanoma is also resistant to a combined treatment characterized by using an inhibitor of BRAF mutation and an inhibitor of immune checkpoint.
  • the melanoma is also resistant to a combined treatment characterized by using an inhibitor of BRAF mutation, an inhibitor of MEK and an inhibitor of immune checkpoint.
  • the subject has or susceptible to have melanoma resistant to at least one of the treatments as described above.
  • the subject having a melanoma resistant is identified by standard criteria.
  • the standard criteria for resistance for example, are Response Evaluation Criteria In Solid Tumors (RECIST) criteria, published by an international consortium including NCI.
  • administering refers to the act of injecting or otherwise physically delivering a substance as it exists outside the body (e.g., an activator of LYN) into the subject, such as by mucosal, intradermal, intravenous, subcutaneous, intramuscular delivery and/or any other method of physical delivery described herein or known in the art.
  • a disease, or a symptom thereof is being treated, administration of the substance typically occurs after the onset of the disease or symptoms thereof.
  • administration of the substance typically occurs before the onset of the disease or symptoms thereof.
  • a “therapeutically effective amount” is meant a sufficient amount of activator of LYN for use in a method for the treatment of melanoma at a reasonable benefit/risk ratio applicable to any medical treatment. It will be understood that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including the age, body weight, general health, sex and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific polypeptide employed; and like factors well known in the medical arts.
  • the daily dosage of the products may be varied over a wide range from 0.01 to 1,000 mg per adult per day.
  • the compositions contain 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, 250 and 500 mg of the active ingredient for the symptomatic adjustment of the dosage to the subject to be treated.
  • a medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, typically from 1 mg to about 100 mg of the active ingredient.
  • An effective amount of the drug is ordinarily supplied at a dosage level from 0.0002 mg/kg to about 20 mg/kg of body weight per day, especially from about 0.001 mg/kg to 7 mg/kg of body weight per day.
  • the activators of LYN as described above may be combined with pharmaceutically acceptable excipients, and optionally sustained-release matrices, such as biodegradable polymers, to form pharmaceutical compositions.
  • pharmaceutically acceptable excipients such as a carboxylate, ethylene glycol, ethylene glycol, ethylene glycol, ethylene glycol, ethylene glycol, ethylene glycol, ethylene glycol, ethylene glycol, ethylene glycol dimethoxysulfate, ethylene glycol, glycerol, glycerol, glycerol, sorbitol, sorbitol, arate, arate, arate, arate, arate, arate, arate, arate, arate, arate, arate, arate, ethylene glycol, ethylene glycol, ethylene glycol, ethylene glycol, ethylene glycol, ethylene glycol, ethylene glycol, glycerol, ethylene glycol, glycerol,
  • compositions of the present invention for oral, sublingual, subcutaneous, intramuscular, intravenous, transdermal, local or rectal administration can be administered in a unit administration form, as a mixture with conventional pharmaceutical supports, to animals and human beings.
  • Suitable unit administration forms comprise oral-route forms such as tablets, gel capsules, powders, granules and oral suspensions or solutions, sublingual and buccal administration forms, aerosols, implants, subcutaneous, transdermal, topical, intraperitoneal, intramuscular, intravenous, subdermal, transdermal, intrathecal and intranasal administration forms and rectal administration forms.
  • the pharmaceutical compositions contain vehicles which are pharmaceutically acceptable for a formulation capable of being injected.
  • vehicles which are pharmaceutically acceptable for a formulation capable of being injected.
  • These may be in particular isotonic, sterile, saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and the like or mixtures of such salts), or dry, especially freeze-dried compositions which upon addition, depending on the case, of sterilized water or physiological saline, permit the constitution of injectable solutions.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form In all cases, the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • Solutions comprising compounds of the invention as free base or pharmacologically acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the polypeptide (or nucleic acid encoding thereof) can be formulated into a composition in a neutral or salt form.
  • Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
  • the carrier can also be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetables oils.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminium monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active polypeptides in the required amount in the appropriate solvent with several of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • sterile powders for the preparation of sterile injectable solutions
  • the preferred methods of preparation are vacuum drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
  • the formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above, but drug release capsules and the like can also be employed.
  • the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intrap eritoneal administration.
  • sterile aqueous media which can be employed will be known to those of skill in the art in light of the present disclosure.
  • one dosage could be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion. Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject.
  • a further object of the present invention relates to a method of screening a drug suitable for the treatment of melanoma comprising i) providing a test compound and ii) determining the ability of said test compound to activate the activity of LYN.
  • Any biological assay well known in the art could be suitable for determining the ability of the test compound to activate the activity of LYN. Such assay is briefly described above.
  • control substance refers a molecule that is inert or has no activity relating to an ability to modulate a biological activity or expression. It is to be understood that test compounds capable of activating the activity of LYN, as determined using in vitro methods described herein, are likely to exhibit similar modulatory capacity in applications in vivo.
  • the test compound is selected from the group consisting of peptides, petptidomimetics, small organic molecules (such as MLR- 1023), aptamers or nucleic acids.
  • the test compound according to the invention may be selected from a library of compounds previously synthesised, or a library of compounds for which the structure is determined in a database, or from a library of compounds that have been synthesised de novo.
  • the test compound may be selected form small organic molecules.
  • FIGURES are a diagrammatic representation of FIGURES.
  • Figure 1 LYN expression in BRAFi sensitive and resistant melanoma cells.
  • Figure 2 Patients with melanoma expressing high level of LYN display a favorable prognosis. Kaplan-Meier analysis of the TCGA cohort. Patients with a low LYN expression in the tumors have a statistically significant reduced overall survival time (p ⁇ 0.0001, log rank test (Mantel-Cox).
  • FIG. 4 LYN allosteric activator, MLR1023 inhibits the invasive properties of melanoma cells. Quantification of the effect of MLR1023 on migration of different melanoma cells. Note that the cells that do not express LYN (patient#2R and patient#5) were not responsive to MLR1023.
  • the A375 S (Sensitive), A375 R (Resistant), WM9 S (Susceptible) and WM9 R (Resistant) human cell lines are grown under 5% C02 at 37 ° C in DMEM (Dulbecco Minimum Eagle Essential Medium) medium), Glutamax (Gibco) + 10% FCS + 50U / ml Penicillin + 50pg / ml Streptomycin.
  • the cells of patients are isolated from biopsies after digestion with DNase, Dispase and Collagenase and cultured under 5% C02 at 37 ° C in RPMI medium (glucose, GlutaMAX, pyruvate, Invitrogen) 1640 Glutamax + 1% FCS + 50U / ml Penicillin + 50 pg / ml streptomycin.
  • Western Blot Cells exposed to the different experimental conditions are lysed for 20 minutes at 4 ° C in Fisher buffer (50mM Hepes pH 7.4, 150mM NaCl, ImM Orthovanadate, 5mM NaF, 20mM NaPPi, Complete 7X cocktail and 1% TritonX-100).
  • the lysates are then centrifuged (13000 rpm for 10 min at 4 ° C.) and the protein concentration is determined using micro BCA reagents (Thermo scientific), according to the supplier's recommendations by spectrophotometry at 561 nm.
  • the membrane is saturated (lOmM Tris-HCl pH 7.4, 150mM NaCl, ImM EDTA, 0.1% Tween-20, 3% BSA and 0.5% gelatin) for lh at room temperature and incubated 18h at 4 ° C, in the presence of the primary antibody directed against the protein of interest in the solution of saturation Then, after 3 washes of 10 min in washing buffer (10 mM Tris-HCl pH 7.6, 150 mM NaCl, 1% NP40), the membrane is incubated with a secondary antibody coupled to peroxidase (in saturation solution) for lh at room temperature. The membrane is washed again three times for 10 min and then the proteins of interest are revealed by chemoluminescence using an ECL reagent (Amersham Biosciences).
  • the cells are seeded at a rate of 150,000 to 200,000 per ml in 60 mm diameter culture dishes and then transfected 24h later with a siRNA targeting the members of the SRC family and siRNA control (Flexitube, Qiagen) at a final concentration of 50nM with the HiPerfect transfection reagent (Qiagen) for 48 or 72h.
  • Control or LYN encoding adenoviruses were from Vector Biolabs. We seed the melanoma cells in 6-well culture plates (400,000 cells per well in 2 ml of RPMI 7% FCS medium) and then infect them the next day with the adenoviral particles (1 and 3 pi / well of particles at 4.9 * 1010 PFU / ml). After 48 hours of infection and three washes with PBS, the cells are taken up in fresh medium, and used either for gene/protein expression, or migration assays
  • Transwell® inserts consists of an upper chamber and a lower chamber, separated by a porous membrane (8m) (Coming).
  • the upper chamber is inoculated with a suspension of 300,000 cells in 200 m ⁇ of RPMI medium without FCS and treated according to each condition.
  • the lower chamber contains only 600pL of RMPI medium 7% FCS.
  • the cells migrate for 24 hours according to chemotaxis from the upper chamber to the lower chamber through the porous membrane.
  • the Transwell® inserts are then deposited in a 4% PFA solution for 10 min at room temperature in order to fix the cells having migrated. These are then stained with crystal violet at 0.2% for lOmin. After rinsing with PBS, drying and cleaning the upper part of the Transwell to remove cells that have not migrated, the cells are visualized under a microscope and counted using a software IMAGE J.
  • RNAs are extracted using the RNeasy kit (Qiagen).
  • the cell lysates are deposited on micro-columns, washed by several short centrifugation steps and eluted in 30 pi of H20 RNase.
  • a spectrophotometer Nanodrop
  • 1 pg of RNA is retro-transcribed into cDNA with oligo dT as primer, according to the supplier's protocol (Reverse Transcription System, Promega).
  • a 1 / 50th dilution of cDNA is incubated with 100pL of SYBR Green PCR master reagent mix (Applied biosystems) and the primers specific to the genes of interest diluted to 1/20.
  • the PCR reaction is analyzed in real time using a StepOnePlus real time PCR system thermocycler (Applied Biosystems).
  • the relative quantization of the amplicons was performed by the 2 (- AACT) method. Detection of the housekeeping GAPDH gene is used to normalize the results.
  • the primer sequences used for each cDNA are analyzed in real time using a StepOnePlus real time PCR system thermocycler (Applied Biosystems).
  • the relative quantization of the amplicons was performed by the 2 (- AACT) method. Detection of the housekeeping GAPDH gene is used to normalize the results.
  • the primer sequences used for each cDNA is analyzed in real time using a StepOnePlus real time PCR system thermocycler (Applied Biosystems).
  • LYN expression is decreased in BRAFi-resistant melanoma cells.
  • LYN impedes the metastatic properties of melanoma cells.
  • LYN it is clearly involved in the control of metastatic properties of melanoma cells. According to our data, LYN might be a valuable target to prevent or impair melanoma spreading. However, metastasis inhibition would be achieved through an increase in LYN expression or an activation of LYN activity.
  • MLR1023 a LYN kinase activator, MLR1023 is currently in phase 2b clinical trial for type 2 diabetes indication [8] Therefore, this drug, if efficient in preclinical model setting can rapidly be repositioned in the treatment of melanomas.
  • LYN acted negatively on the metastatic properties of melanoma cells.
  • the Lyn kinase activator MLR-1023 is a novel insulin receptor potentiator that elicits a rapid-onset and durable improvement in glucose homeostasis in animal models of type 2 diabetes.

Abstract

La présente invention concerne une méthode et une composition pour le traitement du mélanome. Les inventeurs ont démontré qu'une diminution de l'expression de LYN, dans des cellules de mélanomes et de tumeurs, est liée à l'acquisition d'une résistance à des thérapies ciblées. Les inventeurs ont également montré l'inhibition de la propension métastatique d'une cellule de mélanome par un activateur allostérique de LYN. Plus particulièrement, la présente invention concerne une méthode permettant de prédire, chez un sujet souffrant d'un mélanome, si ledit mélanome est résistant, ou s'il peut devenir résistant. La présente invention concerne en outre des méthodes permettant de traiter un mélanome et un mélanome résistant chez un sujet en ayant besoin, comprenant une étape consistant à administrer audit sujet une quantité thérapeutiquement efficace d'un activateur de LYN.
PCT/EP2019/083926 2018-12-07 2019-12-06 Méthodes et compositions pour le traitement du mélanome WO2020115261A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP18306642.2 2018-12-07
EP18306642 2018-12-07

Publications (1)

Publication Number Publication Date
WO2020115261A1 true WO2020115261A1 (fr) 2020-06-11

Family

ID=64665808

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2019/083926 WO2020115261A1 (fr) 2018-12-07 2019-12-06 Méthodes et compositions pour le traitement du mélanome

Country Status (1)

Country Link
WO (1) WO2020115261A1 (fr)

Citations (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
EP0368684A1 (fr) 1988-11-11 1990-05-16 Medical Research Council Clonage de séquences d'immunoglobulines de domaines variables.
EP0404097A2 (fr) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Récepteurs mono- et oligovalents, bispécifiques et oligospécifiques, ainsi que leur production et application
WO1993011161A1 (fr) 1991-11-25 1993-06-10 Enzon, Inc. Proteines multivalentes de fixation aux antigenes
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO2003063792A2 (fr) 2002-01-30 2003-08-07 The Brigham And Women's Hospital, Inc. Compositions et methodes associees a tim-3, molecule de surface cellulaire specifique a th1
WO2004004771A1 (fr) 2002-07-03 2004-01-15 Ono Pharmaceutical Co., Ltd. Compositions immunostimulantes
WO2004056875A1 (fr) 2002-12-23 2004-07-08 Wyeth Anticorps anti pd-1 et utilisations
US6982321B2 (en) 1986-03-27 2006-01-03 Medical Research Council Altered antibodies
WO2006003388A2 (fr) 2004-06-30 2006-01-12 Domantis Limited Compositions et procedes pour le traitement de troubles inflammatoires
WO2006030220A1 (fr) 2004-09-17 2006-03-23 Domantis Limited Compositions monovalentes pour la liaison au cd40l et procedes d'utilisation
US7087409B2 (en) 1997-12-05 2006-08-08 The Scripps Research Institute Humanization of murine antibody
WO2006121168A1 (fr) 2005-05-09 2006-11-16 Ono Pharmaceutical Co., Ltd. Anticorps monoclonaux humains pour mort programmee 1 (mp-1) et procedes pour traiter le cancer en utilisant des anticorps anti-mp-1 seuls ou associes a d’autres immunotherapies
WO2007005874A2 (fr) 2005-07-01 2007-01-11 Medarex, Inc. Anticorps monoclonaux humains diriges contre un ligand de mort programmee de type 1(pd-l1)
US7432059B2 (en) 2000-06-28 2008-10-07 Dana-Farber Cancer Institute, Inc. Methods of identifying compounds that upmodulate T cell activation in the presence of a PD-1 mediated signal
WO2008156712A1 (fr) 2007-06-18 2008-12-24 N. V. Organon Anticorps dirigés contre le récepteur humain de mort programmée pd-1
WO2009054864A1 (fr) 2007-10-26 2009-04-30 Rigel Pharmaceuticals, Inc. Triazoles substitués par aryle polycyclique et triazoles substitués par hétéroaryle polycyclique utiles comme inhibiteurs d'axl
WO2009114335A2 (fr) 2008-03-12 2009-09-17 Merck & Co., Inc. Protéines de liaison avec pd-1
WO2010003055A1 (fr) 2008-07-03 2010-01-07 Melior Pharmaceuticals I, Inc. Composés et procédés de traitement de troubles liés au métabolisme du glucose
WO2010036959A2 (fr) 2008-09-26 2010-04-01 Dana-Farber Cancer Institute Anticorps anti-pd-1, pd-l1, et pd-l2 humains et leurs utilisations
WO2010077634A1 (fr) 2008-12-09 2010-07-08 Genentech, Inc. Anticorps anti-pd-l1 et leur utilisation pour améliorer la fonction des lymphocytes t
WO2010089411A2 (fr) 2009-02-09 2010-08-12 Universite De La Mediterranee Anticorps pd-1 et anticorps pd-l1 et leurs utilisations
WO2010117057A1 (fr) 2009-04-10 2010-10-14 協和発酵キリン株式会社 Procédé pour le traitement d'une tumeur sanguine utilisant un anticorps anti-tim-3
WO2011066389A1 (fr) 2009-11-24 2011-06-03 Medimmmune, Limited Agents de liaison ciblés dirigés contre b7-h1
WO2011082400A2 (fr) 2010-01-04 2011-07-07 President And Fellows Of Harvard College Modulateurs du récepteur immunosuppresseur pd-1 et procédés d'utilisation de ceux-ci
WO2011110621A1 (fr) 2010-03-11 2011-09-15 Ucb Pharma, S.A. Produits biologiques : anticorps anti-pd-1 agonistes humanisés
WO2011155607A1 (fr) 2010-06-11 2011-12-15 協和発酵キリン株式会社 Anticorps anti-tim-3
WO2012018857A2 (fr) * 2010-08-02 2012-02-09 The Broad Institute Of Mit And Harvard Prédiction et suivi de la réponse à une thérapie anticancéreuse sur la base du profilage de l'expression génique
WO2013006490A2 (fr) 2011-07-01 2013-01-10 Cellerant Therapeutics, Inc. Anticorps se liant spécifiquement à tim3
WO2013079174A1 (fr) 2011-11-28 2013-06-06 Merck Patent Gmbh Anticorps anti-pd-l1 et utilisations associées
US8617546B2 (en) 2008-10-02 2013-12-31 Seoul National University Industry Foundation Anticancer agent comprising anti-PD-1 antibody or anti-PD-L1 antibody
WO2014053650A1 (fr) * 2012-10-04 2014-04-10 Ab Science Utilisation de masitinib pour le traitement du cancer chez des sous-populations de patients identifiées à l'aide de facteurs de prédiction
WO2014055648A1 (fr) 2012-10-02 2014-04-10 Bristol-Myers Squibb Company Combinaison d'anticorps anti-kir et d'anticorps anti-pd-1 pour le traitement du cancer
WO2014055897A2 (fr) 2012-10-04 2014-04-10 Dana-Farber Cancer Institute, Inc. Anticorps monoclonaux humains anti pd-l1 et procédés d'utilisation
WO2014150677A1 (fr) 2013-03-15 2014-09-25 Bristol-Myers Squibb Company Inhibiteurs de l'indoléamine 2,3-dioxygénase (ido)
WO2014194302A2 (fr) 2013-05-31 2014-12-04 Sorrento Therapeutics, Inc. Protéines de liaison à l'antigène qui se lient à pd-1
WO2014195852A1 (fr) 2013-06-03 2014-12-11 Glaxosmithkline Intellectual Property (No.2) Limited Combinaisons d'un anticorps anti-pd-l1 et d'un inhibiteur de mek et/ou d'un inhibiteur de braf
WO2015035606A1 (fr) 2013-09-13 2015-03-19 Beigene, Ltd. Anticorps anti-pd1 et leur utilisation comme produits thérapeutiques et produits de diagnostic
WO2015048520A1 (fr) 2013-09-27 2015-04-02 Genentech, Inc. Formulations d'anticorps anti-pdl1
WO2015117002A1 (fr) 2014-01-31 2015-08-06 Novartis Ag Molécules d'anticorps anti-tim-3 et leurs utilisations
US20150366867A1 (en) * 2012-12-20 2015-12-24 Alan R. Tall Methods of treating atherosclerosis or myeloproliferative neoplasms by administering a lyn kinase activator

Patent Citations (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US6982321B2 (en) 1986-03-27 2006-01-03 Medical Research Council Altered antibodies
EP0368684A1 (fr) 1988-11-11 1990-05-16 Medical Research Council Clonage de séquences d'immunoglobulines de domaines variables.
EP0404097A2 (fr) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Récepteurs mono- et oligovalents, bispécifiques et oligospécifiques, ainsi que leur production et application
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO1993011161A1 (fr) 1991-11-25 1993-06-10 Enzon, Inc. Proteines multivalentes de fixation aux antigenes
US7087409B2 (en) 1997-12-05 2006-08-08 The Scripps Research Institute Humanization of murine antibody
US7432059B2 (en) 2000-06-28 2008-10-07 Dana-Farber Cancer Institute, Inc. Methods of identifying compounds that upmodulate T cell activation in the presence of a PD-1 mediated signal
US7709214B2 (en) 2000-06-28 2010-05-04 Dana-Farber Cancer Institute, Inc. Methods for upregulating an immune response with agents that inhibit the intereaction between PD-L2 and PD-1
WO2003063792A2 (fr) 2002-01-30 2003-08-07 The Brigham And Women's Hospital, Inc. Compositions et methodes associees a tim-3, molecule de surface cellulaire specifique a th1
WO2004004771A1 (fr) 2002-07-03 2004-01-15 Ono Pharmaceutical Co., Ltd. Compositions immunostimulantes
WO2004056875A1 (fr) 2002-12-23 2004-07-08 Wyeth Anticorps anti pd-1 et utilisations
WO2006003388A2 (fr) 2004-06-30 2006-01-12 Domantis Limited Compositions et procedes pour le traitement de troubles inflammatoires
WO2006030220A1 (fr) 2004-09-17 2006-03-23 Domantis Limited Compositions monovalentes pour la liaison au cd40l et procedes d'utilisation
WO2006121168A1 (fr) 2005-05-09 2006-11-16 Ono Pharmaceutical Co., Ltd. Anticorps monoclonaux humains pour mort programmee 1 (mp-1) et procedes pour traiter le cancer en utilisant des anticorps anti-mp-1 seuls ou associes a d’autres immunotherapies
WO2007005874A2 (fr) 2005-07-01 2007-01-11 Medarex, Inc. Anticorps monoclonaux humains diriges contre un ligand de mort programmee de type 1(pd-l1)
WO2008156712A1 (fr) 2007-06-18 2008-12-24 N. V. Organon Anticorps dirigés contre le récepteur humain de mort programmée pd-1
WO2009054864A1 (fr) 2007-10-26 2009-04-30 Rigel Pharmaceuticals, Inc. Triazoles substitués par aryle polycyclique et triazoles substitués par hétéroaryle polycyclique utiles comme inhibiteurs d'axl
WO2009114335A2 (fr) 2008-03-12 2009-09-17 Merck & Co., Inc. Protéines de liaison avec pd-1
WO2010003055A1 (fr) 2008-07-03 2010-01-07 Melior Pharmaceuticals I, Inc. Composés et procédés de traitement de troubles liés au métabolisme du glucose
WO2010036959A2 (fr) 2008-09-26 2010-04-01 Dana-Farber Cancer Institute Anticorps anti-pd-1, pd-l1, et pd-l2 humains et leurs utilisations
US8552154B2 (en) 2008-09-26 2013-10-08 Emory University Anti-PD-L1 antibodies and uses therefor
US8617546B2 (en) 2008-10-02 2013-12-31 Seoul National University Industry Foundation Anticancer agent comprising anti-PD-1 antibody or anti-PD-L1 antibody
WO2010077634A1 (fr) 2008-12-09 2010-07-08 Genentech, Inc. Anticorps anti-pd-l1 et leur utilisation pour améliorer la fonction des lymphocytes t
WO2010089411A2 (fr) 2009-02-09 2010-08-12 Universite De La Mediterranee Anticorps pd-1 et anticorps pd-l1 et leurs utilisations
WO2010117057A1 (fr) 2009-04-10 2010-10-14 協和発酵キリン株式会社 Procédé pour le traitement d'une tumeur sanguine utilisant un anticorps anti-tim-3
WO2011066389A1 (fr) 2009-11-24 2011-06-03 Medimmmune, Limited Agents de liaison ciblés dirigés contre b7-h1
WO2011082400A2 (fr) 2010-01-04 2011-07-07 President And Fellows Of Harvard College Modulateurs du récepteur immunosuppresseur pd-1 et procédés d'utilisation de ceux-ci
WO2011110621A1 (fr) 2010-03-11 2011-09-15 Ucb Pharma, S.A. Produits biologiques : anticorps anti-pd-1 agonistes humanisés
WO2011155607A1 (fr) 2010-06-11 2011-12-15 協和発酵キリン株式会社 Anticorps anti-tim-3
WO2012018857A2 (fr) * 2010-08-02 2012-02-09 The Broad Institute Of Mit And Harvard Prédiction et suivi de la réponse à une thérapie anticancéreuse sur la base du profilage de l'expression génique
WO2013006490A2 (fr) 2011-07-01 2013-01-10 Cellerant Therapeutics, Inc. Anticorps se liant spécifiquement à tim3
WO2013079174A1 (fr) 2011-11-28 2013-06-06 Merck Patent Gmbh Anticorps anti-pd-l1 et utilisations associées
WO2014055648A1 (fr) 2012-10-02 2014-04-10 Bristol-Myers Squibb Company Combinaison d'anticorps anti-kir et d'anticorps anti-pd-1 pour le traitement du cancer
WO2014053650A1 (fr) * 2012-10-04 2014-04-10 Ab Science Utilisation de masitinib pour le traitement du cancer chez des sous-populations de patients identifiées à l'aide de facteurs de prédiction
WO2014055897A2 (fr) 2012-10-04 2014-04-10 Dana-Farber Cancer Institute, Inc. Anticorps monoclonaux humains anti pd-l1 et procédés d'utilisation
US20150366867A1 (en) * 2012-12-20 2015-12-24 Alan R. Tall Methods of treating atherosclerosis or myeloproliferative neoplasms by administering a lyn kinase activator
WO2014150677A1 (fr) 2013-03-15 2014-09-25 Bristol-Myers Squibb Company Inhibiteurs de l'indoléamine 2,3-dioxygénase (ido)
WO2014194302A2 (fr) 2013-05-31 2014-12-04 Sorrento Therapeutics, Inc. Protéines de liaison à l'antigène qui se lient à pd-1
WO2014195852A1 (fr) 2013-06-03 2014-12-11 Glaxosmithkline Intellectual Property (No.2) Limited Combinaisons d'un anticorps anti-pd-l1 et d'un inhibiteur de mek et/ou d'un inhibiteur de braf
WO2015035606A1 (fr) 2013-09-13 2015-03-19 Beigene, Ltd. Anticorps anti-pd1 et leur utilisation comme produits thérapeutiques et produits de diagnostic
WO2015048520A1 (fr) 2013-09-27 2015-04-02 Genentech, Inc. Formulations d'anticorps anti-pdl1
WO2015117002A1 (fr) 2014-01-31 2015-08-06 Novartis Ag Molécules d'anticorps anti-tim-3 et leurs utilisations

Non-Patent Citations (18)

* Cited by examiner, † Cited by third party
Title
"Genbank", Database accession no. NP_002341.1
AHMED, F. ET AL.: "Microenvironment-Driven Dynamic Heterogeneity and Phenotypic Plasticity as a Mechanism of Melanoma Therapy Resistance", FRONTIERS IN ONCOLOGY, vol. 8, 2018, pages 173
BRICKELL, P.M.: "The p60c-src family of protein-tyrosine kinases: structure, regulation, and function", CRITICAL REVIEWS IN ONCOGENESIS, vol. 3, 1992, pages 401 - 446
EVAN INGLEY: "Functions of the Lyn tyrosine kinase in health and disease", CELL COMMUNICATION AND SIGNALING, vol. 10, no. 1, 17 July 2012 (2012-07-17), BIOMED CENTRAL, LONDON, GB, pages 21, XP021133103, ISSN: 1478-811X, DOI: 10.1186/1478-811X-10-21 *
GIROTTI, M.R. ET AL.: "Inhibiting EGF receptor or SRC family kinase signaling overcomes BRAF inhibitor resistance in melanoma", CANCER DISCOV, vol. 3, 2013, pages 158 - 167, XP055145699, DOI: 10.1158/2159-8290.CD-12-0386
GIROTTI, M.R. ET AL.: "Paradox-breaking RAF inhibitors that also target SRC are effective in drug-resistant BRAF mutant melanoma", CANCER CELL, vol. 27, 2015, pages 85 - 96, XP029131331, DOI: 10.1016/j.ccell.2014.11.006
HOHEISEL, NATURE REVIEWS, GENETICS, vol. 7, 2006, pages 200 - 210
HUGO, W. ET AL.: "Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma", CELL, vol. 168, 2017, pages 542
KIM, Y. J. ET AL.,: "Abstract 2785: LYN is a new prognostic and therapeutic target in non-small cell lung cancer", CANCER RESEARCH, vol. 74, no. 19 Suppl., 1 October 2014 (2014-10-01), XP002789294, DOI: 10.1158/1538-7445.AM2014-2785 *
LONG E. ET AL.: "High expression of TRF2, SOX10, and CD10 in circulating tumor microemboli detected in metastatic melanoma patients", A POTENTIAL IMPACT FOR THE ASSESSMENT OF DISEASE AGGRESSIVENESS. CANCER MEDICINE, vol. 5, no. 6, 2016, pages 1022 - 1030
LONG ET AL., CANCER MEDECINE, vol. 5, no. 6, 2016, pages 1022 - 1030
LUKE, J.J. ET AL.: "Targeted agents and immunotherapies: optimizing outcomes in melanoma", NAT REV CLIN ONCOL, vol. 14, 2017, pages 463 - 482
M. S. SAPORITO ET AL: "MLR-1023 Is a Potent and Selective Allosteric Activator of Lyn Kinase In Vitro That Improves Glucose Tolerance In Vivo", JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS, vol. 342, no. 1, 1 July 2012 (2012-07-01), pages 15 - 22, XP055562815, DOI: 10.1124/jpet.112.192096 *
MELLMAN ET AL., NATURE, vol. 480, 2011, pages 480 - 489
OCHMAN, A.R. ET AL.: "The Lyn kinase activator MLR-1023 is a novel insulin receptor potentiator that elicits a rapid-onset and durable improvement in glucose homeostasis in animal models of type 2 diabetes", THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS, vol. 342, 2012, pages 23 - 32, XP055543757, DOI: 10.1124/jpet.112.192187
PARDOLL, NATURE REV CANCER, vol. 12, 2012, pages 252 - 264
QIANQIAN ZHANG ET AL: "Lyn Kinase Promotes the Proliferation of Malignant Melanoma Cells through Inhibition of Apoptosis and Autophagy via the PI3K/Akt Signaling Pathway", JOURNAL OF CANCER, vol. 10, no. 5, 29 January 2019 (2019-01-29), AU, pages 1197 - 1208, XP055562828, ISSN: 1837-9664, DOI: 10.7150/jca.28908 *
VERGANI, E. ET AL.: "Identification of MET and SRC activation in melanoma cell lines showing primary resistance to PLX4032", NEOPLASIA, vol. 13, 2011, pages 1132 - 1142, XP055081482, DOI: 10.1593/neo.111102

Similar Documents

Publication Publication Date Title
CN105324491B (zh) 用于癌症转移的诊断和治疗的方法
JP6212107B2 (ja) 脱毛障害を処置するための方法
KR102226219B1 (ko) 폐암 전이의 진단, 예후 및 치료 방법
CN105980576B (zh) 用于源自乳腺癌的骨转移癌的预后和治疗的方法
CN108192972B (zh) 用于乳腺癌转移的诊断、预后和治疗的方法
CN109790582B (zh) 基于c-maf状态的乳腺癌治疗性治疗
US11674182B2 (en) Biomarker for HER2-positive cancer and anti-HER2 therapy and applications thereof
US20200199689A1 (en) Method for Predicting Effectiveness of Angiogenesis Inhibitor
JP2016518815A (ja) 転移性がんの診断、予後、および処置の方法
US20210186982A1 (en) Methods and compositions for treating melanoma
WO2021048292A1 (fr) Méthodes et compositions pour le traitement d'un mélanome
KR20220031069A (ko) 항-종양 치료의 효과를 진단하기 위한 방법
EP3516071B1 (fr) Procédé et compositions pharmaceutiques de traitement du cancer du poumon
EP3591050B1 (fr) Utilisation de rhoa dans le diagnostic du cancer et le dépistage d'inhibiteurs
US20210072244A1 (en) Methods and compositions for treating melanoma resistant
EP3289104A2 (fr) Procédé pour le traitement de gliomes de haut grade
WO2020115261A1 (fr) Méthodes et compositions pour le traitement du mélanome
KR101514877B1 (ko) APE/Ref -1 및 JAG1/Notch의 대장암 진단용 마커로서의 용도
KR102074559B1 (ko) 위암의 항암제 내성 진단용 바이오마커 및 이의 용도
US20120231020A1 (en) Markers for selecting personalized therapies for the treatment of cancer
US20220364184A1 (en) Compositions and methods for treatment of a poor prognosis subtype of colorectal cancer
US20190055565A1 (en) Prevention, diagnosis and treatment of cancer overexpressing gpr160
WO2012034076A2 (fr) Etv1 à titre de cible diagnostique, pronostique et thérapeutique pour les tumeurs stromales gastro-intestinales
WO2022244807A1 (fr) Gène de fusion de la ltk
EP3067369A1 (fr) Procédés et compositions pour le traitement d'un cancer résistant anti-angiogénique

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: 19813032

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: 19813032

Country of ref document: EP

Kind code of ref document: A1