US20190292529A1 - Novel isoform of anaplastic lymphoma kinase and its uses - Google Patents

Novel isoform of anaplastic lymphoma kinase and its uses Download PDF

Info

Publication number
US20190292529A1
US20190292529A1 US16/258,477 US201916258477A US2019292529A1 US 20190292529 A1 US20190292529 A1 US 20190292529A1 US 201916258477 A US201916258477 A US 201916258477A US 2019292529 A1 US2019292529 A1 US 2019292529A1
Authority
US
United States
Prior art keywords
alk
cancer
talk
ati
cell
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US16/258,477
Inventor
Ping Chi
Thomas Wiesner
Yu Chen
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Memorial Sloan Kettering Cancer Center
Original Assignee
Memorial Sloan Kettering Cancer Center
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 Memorial Sloan Kettering Cancer Center filed Critical Memorial Sloan Kettering Cancer Center
Priority to US16/258,477 priority Critical patent/US20190292529A1/en
Publication of US20190292529A1 publication Critical patent/US20190292529A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • 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
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/542Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/545Compounds containing 5-thia-1-azabicyclo [4.2.0] octane ring systems, i.e. compounds containing a ring system of the formula:, e.g. cephalosporins, cefaclor, or cephalexine
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/10Protein-tyrosine kinases (2.7.10)
    • 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
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • 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
    • C12Q2600/158Expression 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)
    • 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
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the present invention relates to a Truncated isoform of Anaplastic Lymphoma Kinase (“TALK”). Expression of this isoform is associated with malignancy and with responsiveness to ALK inhibitors. Detection of the isoform may be used in diagnostic and therapeutic methods. Because it arises as a result of variant transcription rather than genetic rearrangement, its presence would be undetected by genomic testing.
  • TALK Anaplastic Lymphoma Kinase
  • the present invention relates to a truncated isoform of anaplastic lymphoma kinase, also referred to as “TALK” herein, and its use in diagnosis and treatment of cancer patients. It is based, at least in part, on the discovery of a novel oncogenic TALK, termed “ALK ATI ”, that arose as a result of the establishment of a de novo alternative transcriptional initiation (ATI) site in ALK intron 19 independent of genetic aberrations at the ALK locus.
  • ALK ATI transcript encodes three proteins with a molecular weight of 61.1, 60.8 and 58.7 kDa consisting primarily of the intracellular tyrosine kinase domain.
  • ALK ATI was found to stimulate multiple oncogenic signaling pathways, drive growth factor-independent cell proliferation in vitro, and promote tumourigenesis in vivo.
  • ALK inhibitors were found to suppress the kinase activity of ALK ATI , suggesting that patients with ALK ATI -expressing tumors may benefit from ALK inhibitors.
  • Expression of ALK ATI was found in more than 10% ( ⁇ 11%) of melanomas and sporadically in other cancer types, but not in normal tissues.
  • Detection of TALK e.g., ALK ATI in a cell of a subject may be used to diagnose a cancer in the subject and may be used to determine whether treatment of the subject with an ALK inhibitor would be more likely to confer therapeutic benefit. It is an advantage of the invention that a subject expressing TALK who would appear normal by genomic testing may be identified by detecting TALK transcription, mRNA or protein.
  • the present invention provides an isolated nucleic acid molecule comprising exons 20-29 and a portion but not all of intron 19 of an Anaplastic Lymphoma Kinase (ALK) gene, where the nucleic acid molecule does not comprise exons 1-19 of the ALK gene, either individually or in any combination.
  • ALK gene is a human ALK gene.
  • the portion of intron 19 of the ALK gene is about 400 bp in length located upstream of exon 20 of the ALK gene.
  • the nucleic acid molecule is about 2300-2600 bp in length, such as about 2500 bp in length, e.g., about 2513 bp in length.
  • the nucleic acid molecule is a messenger RNA (mRNA) transcript.
  • the isolated nucleic acid molecule comprises the nucleotide sequence set forth in SEQ ID NO: 11 or a sequence that is at least about 95% homologous thereto.
  • the present invention also provides an isolated nucleic acid molecule consisting essentially of exons 20-29 and a portion but not all of intron 19 of an Anaplastic Lymphoma Kinase (ALK) gene, wherein the nucleic acid molecule does not comprise exons 1-19 of the ALK gene, either individually or in any combination.
  • the isolated nucleic acid molecule consists essentially of the nucleotide sequence set forth in SEQ ID NO: 11 or a sequence that is at least about 95% homologous thereto.
  • the present invention further provides an isolated complementary DNA (cDNA) molecule comprising exons 20-29 of an Anaplastic Lymphoma Kinase (ALK) gene, wherein the cDNA molecule does not comprise exons 1-19 of the ALK gene, either individually or in any combination.
  • ALK Anaplastic Lymphoma Kinase
  • the ALK gene is a human ALK gene.
  • the isolated cDNA molecule comprises nucleic acids 405-2063 of the nucleotide sequence set forth in SEQ ID NO: 11 or a sequence that is at least about 95% homologous thereto.
  • the isolated cDNA molecule comprises nucleic acids 411-2063 of the nucleotide sequence set forth in SEQ ID NO: 11 or a sequence that is at least about 95% homologous thereto. In certain embodiments, the isolated cDNA molecule comprises nucleic acids 465-2063 of the nucleotide sequence set forth in SEQ ID NO: 11 or a sequence that is at least about 95% homologous thereto.
  • the present invention provides an isolated complementary DNA (cDNA) molecule consisting essentially of exons 20-29 of an Anaplastic Lymphoma Kinase (ALK) gene, wherein the cDNA molecule does not comprise exons 1-19 of the ALK gene, either individually or in any combination.
  • cDNA complementary DNA
  • ALK Anaplastic Lymphoma Kinase
  • an isolated cDNA molecule consisting essentially of nucleic acids 405-2063 of the nucleotide sequence set forth in SEQ ID NO: 11 or a sequence that is at least about 95% homologous thereto, wherein the cDNA molecule does not comprise exons 1-19 of the ALK gene, either individually or in any combination.
  • the present invention further provides an isolated cDNA molecule consisting essentially of nucleic acids 411-2063 of the nucleotide sequence set forth in SEQ ID NO: 11 or a sequence that is at least about 95% homologous thereto, wherein the cDNA molecule does not comprise exons 1-19 of the ALK gene, either individually or in any combination.
  • the present invention further provides an isolated cDNA molecule consisting essentially of nucleic acids 465-2063 of the nucleotide sequence set forth in SEQ ID NO: 11 or a sequence that is at least about 95% homologous thereto, wherein the cDNA molecule does not comprise exons 1-19 of the ALK gene, either individually or in any combination.
  • the present invention provides an isolated nucleic acid molecule comprising a nucleotide sequence encoding the amino acid sequence set forth in SEQ ID NO: 12 or a sequence that is at least about 95% homologous thereto; an isolated nucleic acid molecule comprising a nucleotide sequence encoding amino acids 3 to 552 of the amino acid sequence set forth in SEQ ID NO: 12 or a sequence that is at least about 95% homologous thereto; or an isolated nucleic acid molecule comprising a nucleotide sequence encoding amino acids 21 to 552 of the amino acid sequence set forth in SEQ ID NO: 12 or a sequence that is at least about 95% homologous thereto.
  • the present invention also provides a vector comprising the above-described isolated nucleic acid molecule, or the above-described isolated cDNA molecule.
  • the invention provides a host cell comprising such vector.
  • the invention provides an isolated polypeptide encoded by the above-descried nucleic acid molecule, or an isolated polypeptide encoded by the above-described cDNA molecule.
  • the present invention further provides an isolated polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 12 or a sequence that is at least about 95% homologous thereto, where the polypeptide does not comprise the amino acid sequence set forth in SEQ ID NO; 3. Also provided is an isolated polypeptide comprising amino acids 3 to 552 of the amino acid sequence set forth in SEQ ID NO: 12 or a sequence that is at least about 95% homologous thereto, where the polypeptide does not comprise the amino acid sequence set forth in SEQ ID NO; 3.
  • the present invention further provides an isolated polypeptide comprising amino acids 21 to 552 of the amino acid sequence set forth in SEQ ID NO: 12 or a sequence that is at least about 95% homologous thereto, where the polypeptide does not comprise the amino acid sequence set forth in SEQ ID NO; 3.
  • the present invention provides an isolated polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 12 or a sequence that is at least about 95% homologous thereto, where the polypeptide does not comprise the amino acid sequence set forth in SEQ ID NO; 4. Also provided is an isolated polypeptide comprising amino acids 3 to 552 of the amino acid sequence set forth in SEQ ID NO: 12 or a sequence that is at least about 95% homologous thereto, where the polypeptide does not comprise the amino acid sequence set forth in SEQ ID NO; 4.
  • the present invention further provides an isolated polypeptide comprising amino acids 21 to 552 of the amino acid sequence set forth in SEQ ID NO: 12 or a sequence that is at least about 95% homologous thereto, where the polypeptide does not comprise the amino acid sequence set forth in SEQ ID NO; 4.
  • the invention also provides an antibody that binds to any of the above-described polypeptides, wherein the antibody does not bind to a wild type ALK.
  • the wild type ALK is a wild type human ALK, e.g., one comprising the amino acid sequence set forth in SEQ ID NO: 1.
  • the present invention also provides a method of diagnosing a cell as a cancer cell comprising detecting the presence of a Truncated isoform of Anaplastic Lymphoma Kinase (TALK) in the cell, where the presence of the TALK indicates that the cell is a cancer cell.
  • the method comprises determining the presence of a detectable TALK mRNA transcript, a detectable TALK cDNA molecule corresponding thereto, a detectable TALK polypeptide encoded thereby, and/or a detectable alternative transcriptional initiation (ATI) site in intron 19 of an ALK gene in the cell.
  • ATI detectable alternative transcriptional initiation
  • the present invention provides a method of determining whether an anti-cancer effect is likely to be produced in a cancer by an ALK inhibitor, comprising determining whether one or more cell of the cancer contains a detectable TALK, wherein the presence of a detectable TALK in the cell indicates that an ALK inhibitor would have an anti-cancer effect on the cancer.
  • the method comprises determining the presence of a detectable TALK mRNA transcript, a detectable TALK cDNA molecule corresponding thereto, a detectable TALK polypeptide encoded thereby, and/or a detectable ATI site in intron 19 of an ALK gene in the cell.
  • the present invention further provides a method of treating a subject having a cancer comprising: (a) determining whether a subject is likely to obtain therapeutic benefit from an ALK inhibitor, comprising determining whether one or more cancer cell of the subject contains a detectable TALK, wherein the presence of a detectable TALK in the cancer cell of the subject indicates that the subject is likely to benefit from an ALK inhibitor; and (b) treating the subject who is likely to benefit from an ALK inhibitor with a therapeutic amount of an ALK inhibitor.
  • the method comprises treating the subject with an alternative therapy other than an ALK inhibitor if no detectable TALK is present in the cancer cell of the subject.
  • the method further comprises obtaining a sample of one or more cancer cell of the subject before treating the subject with an ALK inhibitor.
  • the subject who is likely to benefit from an ALK inhibitor receives an ALK inhibitor and one or more additional cancer treatment selected from the group consisting of one or more BRAF inhibitor, one or more MEK inhibitor, one or more immunologic inhibitor, one or more CDK4 inhibitor, one or more CDK6 inhibitor, one or more alklyating agent, one or more topoisomerase inhibitor, one or more anti-metabolite, one or more anti-microtubule agent, one or more cytotoxic antibiotic, radiation therapy, chemotherapy, and combinations thereof.
  • the immunologic inhibitor is selected from the group consisting an anti-PD-1 antibody, an anti-CTLA-4 antibody, an anti-PD-L1 antibody.
  • the subject who is likely to benefit from an ALK inhibitor receives an ALK inhibitor, an anti-CTLA-4 antibody, and an anti-PD-1 antibody.
  • the subject further receives radiation and chemotherapy.
  • the method comprises determining the presence of a detectable TALK mRNA transcript, a detectable TALK cDNA molecule, a detectable TALK polypeptide encoded thereby, and/or a detectable ATI site in intron 19 of an ALK gene in the cell.
  • the present invention also provides a kit for determining whether an anti-cancer effect is likely to be produced in a cancer by an ALK inhibitor, comprising a means for determining the presence of a detectable TALK in one or more cell of the cancer.
  • the kit comprises means for determining the level of a TALK mRNA transcript, a TALK cDNA molecule corresponding thereto, and/or a TALK polypeptide encoded thereby.
  • the means for determining the level of a TALK mRNA transcript is selected from the group consisting of probe hybridization, polymerase chain reaction (PCR), Northern blot, sequencing, microarray, and combinations thereof.
  • the kit comprises a nucleic acid probe that hybridizes with the TALK mRNA transcript to determine the level of the TALK mRNA transcript.
  • the probe hybridization is a probe-based NanoString nCounter assay.
  • the kit comprises a color coded probe pair that hybridizes with the TALK mRNA transcript to determine the level of the TALK mRNA transcript.
  • the PCR is selected from the group consisting of reverse transcription polymerase chain reaction (RT-PCR), quantitative reverse transcriptase PCR, real-time PCR, quantitative real-time PCR, and combinations thereof.
  • the kit comprises a pair of nucleic acid primers that hybridizes with the TALK mRNA to determine the level of the TALK mRNA transcript.
  • the means for determining the level of a TALK cDNA molecule is PCR.
  • the kit comprises a pair of nucleic acid primers that hybridize with the TALK cDNA molecule to determine the level of the TALK cDNA molecule.
  • the means for determining the level of a TALK polypeptide is selected from the group consisting of antibody binding, immunohistochemistry, Western blot, a functional assay, enzyme linked immunosorbent assay (ELISA), radioimmunoassays (RIA), enzyme immunoassays (EIA), mass spectrometry, a 1-D or 2-D gel-based analysis system, immunoprecipitation, and combinations thereof.
  • the kit comprises an antibody or a fragment thereof that specifically binds to the TALK polypeptide.
  • the functional assays is a kinase assay.
  • the kit comprises means for determining the presence of an ATI site in intron 19 of an ALK gene in one or more cell of the cancer.
  • the means for determining the presence of an ATI site in intron 19 of an ALK gene is selected from the group consisting of Northern blot, Chromatin immunoprecipitation (ChIP)-seq, ChIP-qPCR, Rapid Amplification of cDNA Ends (RACE)-PCR, and combinations thereof.
  • the present invention provides a means for determining the presence of a detectable TALK for use in a method of determining whether an anti-cancer effect is likely to be produced in a cancer by an ALK inhibitor, the method characterized by determining whether one or more cell of the cancer contains a detectable TALK, where the presence of a detectable TALK in the cell indicates that an ALK inhibitor would have an anti-cancer effect on the cancer.
  • the means is for determining the level of a TALK mRNA transcript in one or more cell of the cancer.
  • the means is selected from the group consisting of probe hybridization, polymerase chain reaction (PCR), Northern blot, sequencing, microarray, and combinations thereof.
  • the probe hybridization is a probe-based NanoString nCounter assay.
  • the PCR is selected from the group consisting of reverse transcription polymerase chain reaction (RT-PCR), quantitative reverse transcriptase PCR, real-time PCR, quantitative real-time PCR, and combinations thereof.
  • the means is for determining the level of a TALK cDNA molecule in one or more cell of the cancer.
  • the means is PCR.
  • the means is for determining the level of a TALK polypeptide in one or more cell of the cancer.
  • the means is selected from the group consisting of antibody binding, immunohistochemistry, Western blot, a functional assay, enzyme linked immunosorbent assay (ELISA), radioimmunoassays (RIA), enzyme immunoassays (EIA), mass spectrometry, a 1-D or 2-D gel-based analysis system, immunoprecipitation, and combinations thereof.
  • the functional assays is a kinase assay.
  • the means is for determining the presence of an ATI site in intron 19 of an ALK gene in one or more cell of the cancer.
  • the means is selected from the group consisting of Northern blot, Chromatin immunoprecipitation (ChIP)-seq, ChIP-qPCR, Rapid Amplification of cDNA Ends (RACE)-PCR, and combinations thereof.
  • the present invention further provides an ALK inhibitor for use in treating a subject having cancer, wherein the ALK inhibitor is more likely to produce an anti-cancer effect if one or more cancer cell of the subject contains a detectable TALK.
  • the cancer is selected from the group consisting of melanoma, thyroid carcinoma, lung adenocarcinoma, lung squamous cell carcinoma, renal clear cell carcinoma, and breast cancer.
  • the cancer is melanoma.
  • the cancer is anaplastic thyroid carcinoma.
  • the ALK inhibitor is selected from the group consisting of crizotinib, ceritinib, NVP-TAE684, alectinib; AP26113, ASP-3026, CEP-37440, NMS-E628, PF-06463922, TSR-011, RXDX-101, and X-396.
  • the ALK inhibitor is crizotinib.
  • the TALK mRNA transcript is the above-described nucleic acid molecule.
  • the TALK cDNA molecule is the above-described cDNA molecule.
  • the TALK polypeptide is the above-described polypeptide.
  • FIGS. 1A-1H Alternative transcriptional initiation (ATI) results in a novel ALK transcript.
  • 1 A Distribution of RNA-seq reads of ALK variant transcripts: ALK ATI RNAseq reads aligned to both ALK intron 19 and exons 20-29; full-length ALK WT RNA-seq reads aligned to all ALK exons, but not to the introns; translocated ALK RNA-seq reads aligned only to ALK exons 20-29.
  • 1 B Mapping of the ATI sites of ALKATI by 5′-RACE and next-generation sequencing.
  • 1 C ChIP-seq profile of H3K4me3 at the ATI site of an ALK ATI -expressing tumor (MM-15), a melanoma cell line (SKMEL-524) without ALK ATI expression, and a lung cancer cell line (H3122) with a EML4-ALK fusion. The shape of the peak shows a slow taper towards the gene body characteristic of active promoters.
  • 1 F Quantitative mRNA profiling of different ALK variants using Nanostring nCounter.
  • Two ALK WT -expressing neuroblastoma cell lines (SK-N-BE2, SK-N-DZ), two lung cancer cell lines (H3122, H2228) with EML4-ALK translocations, nine ALKATI expressing tumors: 8 melanomas (MM) and 1 anaplastic thyroid carcinoma (ATC).
  • FIGS. 2A-2F The ALKATI transcript encodes three shortened ALK proteins containing mainly the ALK kinase domain.
  • ALK, wildtype (ALK WT ) consists of a signaling peptide from amino acid (aa) position 1-18, an extracellular segment with two MAM domains (meprin, A-5 protein, and receptor protein-tyrosine phosphatase mu) and one glycine rich region (Gly), a transmembrane segment, a juxtamembrane segment, and an intracellular segment with the tyrosine kinase domain (kinase, aa 1116-1392).
  • ALK mutations occur usually in the kinase domain, such as ALK F1174L .
  • the EML4-ALK fusion protein and ALK ATI contain the entire kinase domain and parts of the juxtamembrane segment.
  • the translation of ALK ATI initiates at one of three in-frame start codons (ATGs) as indicated.
  • 2 B Immunoblots of total- (t-) and phosphorylated- (p-) ALK in two ALK WT -expressing neuroblastoma cell lines (SK-N-DZ, SK-N-BE2), two lung cancer cell lines (H3122, H2228) expressing different variants of EML4-ALK fusion, three tumor samples expressing ALK ATI (MM-15, MM-74, ATC-28) and a negative control melanoma cell line (SKMEL-28).
  • 2 C Immunoblots of 293T cells transduced with ALK ATI , in which the three predicted start codons were mutated from ATG to AAG, individually or in combination as indicated.
  • 2 D Co-immunoprecipitation (IP) and immunoblots (IB) of the indicated epitopes in 293T cells with exogenous expression of V5-tagged ALK ATI (V5-ALK ATI ) or HA-tagged ALKATI (HA-ALK ATI ), or both, demonstrating that ALK ATI proteins self-interact.
  • 2 E ALK immunofluorescence in NIH-3T3 cells expressing the indicated ALK isoforms. Scale bar, 25 ⁇ m.
  • 2 F HE (haematoxylin-eosin) stain and ALK immunohistochemistry in ALK ATI -expressing human tumor samples. Scale bar, 50 ⁇ m.
  • FIGS. 3A-3C ALK ATI promotes growth factor-independent proliferation in vitro and tumourigenesis in vivo.
  • 3 B Immunoblots to determine the ALK levels in Ba/F3 cells. Cells were previously transformed by the expression of different ALK isoforms and selected for growth in the absence of IL-3. All ALK variants were phosphorylated when expressed at levels required for IL-3-independent growth.
  • FIGS. 4A-4H ALK ATI -expression confers sensitivity to ALK inhibitors in vitro and in vivo.
  • 4 B Representative immunoblots of Ba/F3 cells expressing ALK ATI and treated with increasing concentrations of crizotinib for 2 hours.
  • 4 C Normalised tumor volume over time in SCID mice implanted with NIH-3T3 cells expressing the indicated ALK isoforms and treated with either vehicle or crizotinib (100 mg/kg/day).
  • 4 G Quantitative mRNA profiling of ALK ATI using Nanostring nCounter. Controls expressing ALK WT , EML4-ALK or ALK ATI compared to the melanoma metastasis of patient MM-382. Scale bar, 50 ⁇ m.
  • 4 H Computed tomography (CT) images of a representative subcutaneous melanoma metastasis on the left hip of patient 1 (MM-382) before and after crizotinib treatment, respectively.
  • CT Computed tomography
  • FIGS. 5A-5C Comparison of the RNA seq profiles of various ALK transcript.
  • RNA-seq data are displayed in the Integrative Genomics Viewer (IGV).
  • the grey bars/arrows indicate the sequencing reads.
  • the blue lines connect sequencing reads that are aligned over the splice site of joining exons.
  • 5 A The ALK ATI transcript shows expression of ALK exons 20-29 and of approx. 400 bps in intron 19 (blue shaded area). No expression of exon 1-19 or intronic areas, other than in intron 19, is observed.
  • the detailed view illustrates that the sequencing reads align continuously between exon 20 and intron 19 indicating uninterrupted transcription.
  • the 5′-UTR of ALK ATI (intron 19) and exon 20-29 are expressed at comparable levels.
  • 5 B The full-length ALK WT transcript shows expression of all ALK exons and only very little expression of the introns. The detailed view displays that the sequencing reads align sharply to the exons, but not to the intron 19 region, which is present in ALK ATI (blue shaded area).
  • 5 C The ALK fusion transcript of a non-small cell lung cancer with an EML4-ALK translocation shows expression of ALK exons 20-29, and little expression of exons 1-19 and all introns. The detailed view illustrates that the transcription starts mainly at exon 20 due to the preserved splice site. Only few reads are aligned to the intron 19 region (blue shaded area). The green-labelled reads highlight chimeric read pairs indicating the EML4-ALK translocation.
  • FIGS. 6A-6G Identification of the novel ALK ATI transcript.
  • 6 A IGV view of the 5′-RACE-cDNA fragments obtained by massively parallel sequencing. The vast majority of sequencing reads (grey arrows) start within the main ATI site of 25 base pairs (hg19 chr2:29,446,744-768).
  • 6 B Percentage of reads starting at the ATI site in ALK ATI -expressing tumor samples.
  • 6 C Sanger sequencing of the cloned 5′-RACE-cDNA fragments confirm the continuous transcription starting in ALK intron 19 and extending to exons 20-21.
  • 6 D The ALK ATI transcript consists of approximately 400 bp upstream of exon 20 and of ALK exons 20-29.
  • the transcriptional initiation site was defined as the first base pair at which more than 5% of the transcripts were initiated (chr2:29,446,766). Other major transcriptional initiation sites are marked in red, the 5′- and 3′-UTRs in dark blue, the coding DNA sequence (CDS) in black and in box, and the first and last base of each exon in light blue.
  • the translation is initiated at 3 start codons (ATGs; bold and underlined): 1st ATG: hg19 chr2:29,446,360-2, 2nd ATG (+7-9), and 3rd ATG (+61-3). 6 E, The amino acid sequence of ALK ATI . The translation is initiated at one of 3 start codons.
  • the corresponding 3 methionines result in 3 different proteins, 61.08 kDa (552 amino acids), 60.82 kDa (550 amino acids) and 58.71 kDa (532 amino acids).
  • the kinase domain is highlighted in red letters.
  • the lysine in the ATP binding domain in ALK WT is marked bold and underlined, and was mutated to methionine (p.K1150M) in the kinase-dead ALKATI-KD.
  • RNA-seq Sashimi plot illustrating the shorter V2 and the longer V1 ALK transcripts by the sharp drop of sequencing reads in the 3′ UTR at chr2:29,415,640 for V2 and at chr2:29,413,840 for V1.
  • FIGS. 7A-7D RNApol II and H3K4me3 are enriched at the ATI site of ALK ATI -expressing tumor samples.
  • 7 A Primer sequences used for ChIP-qPCR.
  • 7 B Schematics of the ChIP-qPCR primer binding sites at the ALK ATI locus.
  • 7 C and 7 D ChIP-qPCR of (C) H3K4me3 and (D) RNApol II at the ATI site demonstrating enrichment of both marks in the ALK ATI -expressing human tumor samples, but not in the negative controls, including a lung cancer cell line with EML4-ALK translocation (H3122), and a melanoma cell line (SKMEL-524).
  • FIGS. 8A-8E ALK ATI -expressing tumors in the TCGA dataset.
  • 8 A The frequency of ALK ATI -expressing tumors in more than 5,000 tumor samples from 15 different cancer types in the TCGA RNA-seq dataset.
  • 8 B- 8 E IGV views of the ALK locus of representative ALK ATI -expressing tumor types in the TCGA dataset including B, melanoma; C, lung adenocarcinoma; D, breast invasive carcinoma; E, clear cell renal cell carcinoma. The absence of chimeric sequencing read pairs indicates no translocations in these cases.
  • FIGS. 9A-9D ALK ATI is transcribed from a genomically intact ALK locus.
  • 9 A Interphase FISH with ALK flanking probes demonstrates juxtaposed green and orange signals indicating no ALK rearrangement in MM-15.
  • Scale bar 10 ⁇ m.
  • 9 B Interphase FISH shows no ALK rearrangement, but 3 green/orange fusion signals in the majority of cell nuclei indicating a trisomy 2 in MM-74.
  • Scale bar 10 ⁇ m.
  • the top panel shows the genome-wide array CGH profile of MM-15 with numerous chromosomal gains and losses across the entire genome, which are characteristic of metastatic melanoma. The chromosomes are aligned along the x-axis.
  • the blue line illustrates the relative copy number (log 2 ratio) and the blue bars highlight copy number gains and losses.
  • the middle panel illustrates the relative copy number (blue line) across chromosome 2. Distal to the ALK locus, a loss on the p-arm of chromosome 2 is indicated. The lower panel illustrates the relative copy number across the ALK gene.
  • the red and green squares represent the log 2 ratio of individual aCGH probes (green: positive log 2 ratio; red: negative log 2 ratio). No disruption or selective gains or losses are found at the ALK locus.
  • 9 D The genome-wide array CGH profile of MM-74 shows numerous chromosomal gains and losses across the entire genome in the top panel.
  • the middle panel displays a relative copy number gain of the entire chromosome 2, which is in line with the trisomy of chromosome 2 as indicated by FISH.
  • the lower panel also displays trisomy of chromosome 2 and shows no focal gains and losses at the ALK locus.
  • FIGS. 10A-10D Targeted sequencing and whole-genome sequencing reveals no recurrent genomic aberrations at the ALK locus.
  • 10 A Data from ultra-deep sequencing of the entire ALK locus are displayed in IGV. The genomic region around intron 19 reveals several single nucleotide variations (SNVs). However, the vast majority of SNVs at the ALK locus are also found in the general population as they are detected in the pool of normal DNA, which was used as the control (pooled normal, bottom panel). Numerous SNVs are also documented in The Single Nucleotide Polymorphism database (dbSNP—www.ncbi.nlm.nih.gov/SNP/).
  • FIGS. 11A-11H Local chromatin context at the alternative transcription initiation (ATI) site.
  • 11 A UCSC Genome Browser view at the ATI site.
  • the RepeatMasker track shows transposable elements at the ATI region, including a long-terminal repeat (LTR) in intron 19 (LTR16B2) and a long-interspaced element (LINE) in intron 18.
  • LTR long-terminal repeat
  • LINE long-interspaced element
  • the ENCODE tracks reveal a DNase I hypersensitivity cluster and H3K4me1 enrichment, but no H3K27ac enrichment.
  • 11 B The methylation status of the ALK locus was assessed by custom capture of the entire ALK footprint, followed by bisulfite treatment and next-generation sequencing.
  • Bisulfite sequencing results of H3122 and MM-15 are displayed in the CG-bisulfite mode of IGV.
  • the red color denotes “C” (Cytosine) corresponding to methylated cytosine, which is preserved during the bisulfite reaction.
  • the blue color denotes “T” (thymine) corresponding to un-methylated cytosine, which is converted to uracil in the bisulfite reaction, and subsequently amplified to thymine during PCR.
  • 11 C Methylation level at CpGs in ALK ATI -expressing tumor samples (MM-15 and ATC-28) and non-ALK ATI -expressing control cells (H3122, a lung cancer cell line with EML4-ALK expression and SKMEL-28, a melanoma cell line without ALK ATI expression) at the indicated genomic loci. Black lines mark the LTR16B2 (LTR), red lines mark the LINE. 11 D, Comparison of the methylation status of CpGs adjacent to the ATI site in ALK ATI -expressing tumor samples (MM-15 and ATC-28) and non-ALK ATI -expressing control cells (H3122 and SKMEL-28).
  • the regions flanking LTR16B2 have significantly lower CpG methylation levels in ALK ATI -expressing samples than controls; red dots indicate a statistically significant difference (p ⁇ 0.05) between ALK ATI -expressing and non-expressing samples. Black dots indicate no statistically significant difference.
  • 11 E ChIP-seq profile of H3K27ac at the ALK ATI locus.
  • the 17 blue profiles were retrieved from ENCODE, the 5 red profiles are original data from the inventors' lab. Only the three melanoma samples (MM-15, SKMEL-28, SKMEL-524; bottom), but not the 19 non-melanoma cell lines, show H3K27ac enrichment at the ATI site.
  • 11 G Luciferase reporter assay of LTR16B2 in melanoma cell lines (red) and lung cancer cell lines expressing EML4-ALK (green). In contrast to the lung cancer cell lines with no luciferase activity, melanoma cell lines show low to moderate luciferase activity.
  • 11 H Transcription factor motif analysis of the proximal cis-regulatory region (hg19 chr2: 29,445,000 to 29,447,100).
  • FIGS. 12A-12I ALK ATI is active in vitro, shows nuclear and cytoplasmic localization by immunohistochemistry, and induces tumourigenesis.
  • 12 B Human tumor (MM-15) expressing ALK ATI shows cytoplasmic and nuclear localization of ALK by immunohistochemistry.
  • 12 D Immunoblots of explanted NIH-3T3 tumor grafts expressing various ALK isoforms.
  • ALK ATI was expressed at similar protein levels as in two ALK ATI -expressing human tumor samples.
  • 12 F Immunoblots of explanted melan-a tumor grafts expressing various ALK isoforms in comparison to ALK ATI -expressing human tumor samples.
  • 12 G Flow cytometry analysis of the GFP signal in NIH-3T3 cells stably expressing low (ALK ATI -low) or high levels of ALK ATI (ALK ATI -high) prior to grafting into SCID mice.
  • 12 H Immunoblot of t-ALK in ALK ATI -low and ALK ATI -high cells, confirming differential expression of ATI.
  • 12 I Growth curves of tumor grafts of ALK ATI -low and ALK ATI -high cells.
  • FIGS. 13A-13E Concentration dependent ALK inhibition in ALK ATI , ALK WT -, ALK F1174L -, and EML4-ALK-expressing Ba/F3 cells.
  • FIG. 13 C- 13 E Representative immunoblots of Ba/F3 cells stably expressing (C) ALK WT , (D) ALK F1174L , or (E) EML4-ALK and treated with increasing concentrations of crizotinib for 2 hours. Immunoblots for ALK ATI are shown in FIG. 4B .
  • FIGS. 14A-14F Expression of ALK WT , ALK F1174L , and EML4-ALK confers sensitivity to the ALK inhibitor, crizotinib, in vivo.
  • 14 A- 14 C Bioluminescence of luciferase-labelled NIH-3T3 grafted tumors expressing (A) ALK WT , (B) ALK F1174L , or (C) EML4-ALK over time in SCID mice treated with either vehicle or crizotinib (100 mg/kg/day). Data for ALK ATI -expressing tumors are shown in FIG. 4E .
  • the term “about” or “approximately” means within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, i.e., the limitations of the measurement system. For example, “about” can mean within 3 or more than 3 standard deviations, per the practice in the art. Alternatively, “about” can mean a range of up to 20%, preferably up to 10%, more preferably up to 5%, and more preferably still up to 1% of a given value. Alternatively, particularly with respect to biological systems or processes, the term can mean within an order of magnitude, preferably within 5-fold, and more preferably within 2-fold, of a value.
  • the term “antibody” means not only intact antibody molecules, but also fragments of antibody molecules that retain immunogen-binding ability. Such fragments are also well known in the art and are regularly employed both in vitro and in vivo. Accordingly, as used herein, the term “antibody” means not only intact immunoglobulin molecules but also the well-known active fragments F(ab′)2, and Fab. F(ab′)2, and Fab fragments that lack the Fc fragment of intact antibody, clear more rapidly from the circulation, and may have less non-specific tissue binding of an intact antibody (Wahl et al., J. Nucl. Med. 24:316-325 (1983).
  • the antibodies of the invention comprise whole native antibodies, bispecific antibodies; chimeric antibodies; Fab, Fab′, single chain V region fragments (scFv), fusion polypeptides, humanized antibodies derived from a non-human antibody, and unconventional antibodies.
  • vector refers to any genetic element, such as a plasmid, phage, transposon, cosmid, chromosome, virus, virion, etc., which is capable of replication when associated with the proper control elements and which can transfer gene sequences into cells.
  • vector includes cloning and expression vehicles, as well as viral vectors and plasmid vectors.
  • expression vector refers to a recombinant nucleic acid sequence, e.g., a recombinant DNA molecule, containing a desired coding sequence and appropriate nucleic acid sequences necessary for the expression of the operably linked coding sequence in a particular host organism.
  • Nucleic acid sequences necessary for expression in prokaryotes usually include a promoter, an operator (optional), and a ribosome binding site, often along with other sequences.
  • Eukaryotic cells are known to utilize promoters, enhancers, and termination and polyadenylation signals.
  • treating refers to clinical intervention in an attempt to alter the disease course of the individual or cell being treated, and can be performed either for prophylaxis or during the course of clinical pathology.
  • Therapeutic effects of treatment include, without limitation, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastases, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis.
  • a treatment can prevent deterioration due to a disorder in an affected or diagnosed subject or a subject suspected of having the disorder, but also a treatment may prevent the onset of the disorder or a symptom of the disorder in a subject at risk for the disorder or suspected of having the disorder.
  • an “effective amount” is an amount sufficient to affect a beneficial or desired clinical result upon treatment.
  • An effective amount can be administered to a subject in one or more doses.
  • an effective amount is an amount that is sufficient to palliate, ameliorate, stabilize, reverse or slow the progression of the disease (e.g., a cancer), or otherwise reduce the pathological consequences of the disease (e.g., a cancer).
  • the effective amount is generally determined by the physician on a case-by-case basis and is within the skill of one in the art. Several factors are typically taken into account when determining an appropriate dosage to achieve an effective amount. These factors include age, sex and weight of the subject, the condition being treated, the severity of the condition and the form and effective concentration of the immunoresponsive cells administered.
  • the term “subject” refers to any animal (e.g., a mammal), including, but not limited to, humans, non-human primates, rodents, and the like (e.g., which is to be the recipient of a particular treatment, or from whom cells are harvested).
  • an anti-cancer effect means one or more of a reduction in aggregate cancer cell mass, a reduction in cancer cell growth rate, a reduction in cancer cell proliferation, a reduction in tumor mass, a reduction in tumor volume, a reduction in tumor cell proliferation, a reduction in tumor growth rate, a reduction in tumor metastasis and/or an increase in the proportion of senescent cancer cells.
  • the human anaplastic lymphoma kinase (ALK) gene is located at chr 2p23, contains 29 exons, and encodes a 1,620 amino acid, 220 kDa classical insulin superfamily tyrosine kinase.
  • the mature human ALK protein undergoes post-translational N-linked glycosylation and consists of an extracellular ligand-binding domain, a transmembrane domain, and a single intracellular tyrosine kinase domain.
  • TALK may be used to refer to the truncated isoforms of ALK referred to herein and the mRNA and corresponding cDNA molecule encoding them.
  • the present invention provides for an ALK isoform comprising exons 20-29 but lacking the transmembrane domain and/or extracellular domain.
  • the TALK arises as a result of the establishment of a de novo alternative transcriptional initiation (“ATI”) site rather than a genomic rearrangement or a genomic aberration at the ALK locus.
  • ATI de novo alternative transcriptional initiation
  • the TALK is termed as ALK ATI .
  • the ALK isoform is not comprised in fusion with a portion of another native protein, for example arising from a translocation event, for example, where the 3′ portion of ALK is fused to a portion of the ATIC, C2orf44, CARS, CLTC, EML4, FN1, KIF5B, KLC1, MSN, NPM1, PPFIBP1, RANBP2, SEC31A, SQSTM1, STRN, TFG, TPM3, TPM4, or VCL protein (see the COMIC database, http://cancer.sanger.ac.uk/cancergenome/projects/cosmic/)
  • the present invention provides for an ALK isoform encoded by a mRNA transcript comprising exons 20-29 and a portion but not all of intron 19, but not exons comprised in the transmembrane domain and/or extracellular domain of ALK.
  • the present invention provides for an ALK isoform encoded by a mRNA transcript comprising exons 20-29 and a portion but not all of intron 19, but not exons 1-19, of the ALK gene, either individually or in any combination.
  • the present invention provides for a nucleic acid molecule comprising the transcript.
  • the size of the transcript is about 2300-2600 bases, or about 2500 bases, or about 2513 bases.
  • Certain non-limiting embodiments of the transcript comprise about 400 bases of intron 19 upstream of exon 20.
  • Certain non-limiting embodiments of the invention provide for a cDNA molecule corresponding to said mRNA transcript.
  • Said cDNA molecule may optionally be operably linked to a promoter and/or incorporated into a nucleic acid vector.
  • Said promoter may be positioned directly before the coding sequence.
  • the promoter is a heterologous promoter (that is to say, not the ALK promoter).
  • said mRNA transcript or cDNA molecule lacks nucleic acid sequence encoding at least a 10 or 20 amino acid long fragment of a protein other than ALK.
  • the ALK is human ALK.
  • the corresponding human ALK gene has a sequence as provided by NCBI Reference Sequence: NG_009445.1 Accession No. NG_009445 or an allelic variant thereof.
  • the human ALK protein has the amino acid sequence provided by NCBI Reference Sequence: NP_004295.2 Accession No. NP_004295 (provided below) or an allelic variant thereof.
  • the untruncated or wild type human ALK mRNA has the nucleotide sequence provided by NCBI Reference Sequence: NM_004304.4 Accession No. NM_004304 (provided below) or an allelic variant thereof.
  • the present invention provides for an isolated nucleic acid molecule comprising the nucleic acid sequence set forth in SEQ ID NO: 11 as shown in FIG. 6D , or a sequence that is at least 80%, at least 85%, at least 90%, or at least 95% homologous thereto, including sequences that are not identical to SEQ ID NO: 11, but lacking sequence encoding exons 1-19 of ALK, either individually or in any combination.
  • the nucleic acid molecule comprises a nucleotide sequence encoding the amino acid sequence set forth in SEQ ID NO: 12 as shown in FIG. 6E , or a sequence that is at least 80%, at least 85%, at least 90%, or at least 95% homologous thereto, including sequences that are not identical to SEQ ID NO: 12.
  • the nucleic acid molecule comprises a nucleotide sequence encoding an amino acid sequence that comprises at least one (e.g., no more than 2, no more than 3, no more than 4, no more than 5, no more than 6, no more than 7, no more than 8, no more than 9, no more than 10, no more than 11, no more than 12, no more than 13, no more than 14, no more than 15, no more than 16, no more than 17, no more than 18, no more than 19, or no more than 20) amino acid variation relative to SEQ ID NO: 12, where the resulting sequence is not found in a naturally occurring protein, e.g., a wild-type protein.
  • the nucleic acid molecule comprises a nucleotide sequence encoding amino acids 3-552 of SEQ ID NO: 12 or a sequence that is at least 80%, at least 85%, at least 90%, or at least 95% homologous thereto, including sequences that are not identical to amino acids 3-552 of SEQ ID NO: 12.
  • the nucleic acid molecule comprises a nucleotide sequence encoding an amino acid sequence that comprises at least one (e.g., no more than 2, no more than 3, no more than 4, no more than 5, no more than 6, no more than 7, no more than 8, no more than 9, no more than 10, no more than 11, no more than 12, no more than 13, no more than 14, no more than 15, no more than 16, no more than 17, no more than 18, no more than 19, or no more than 20) amino acid variation relative to amino acids 3-552 of SEQ ID NO: 12, where the resulting sequence is not found in a naturally occurring protein, e.g., a wild-type protein.
  • the nucleic acid molecule comprises a nucleotide sequence encoding amino acids 21-552 of SEQ ID NO: 12 or a sequence that is at least 80%, at least 85%, at least 90%, or at least 95% homologous thereto, including sequences that are not identical to amino acids 21-552 of SEQ ID NO: 12.
  • the nucleic acid molecule comprises a nucleotide sequence encoding an amino acid sequence that comprises at least one (e.g., no more than 2, no more than 3, no more than 4, no more than 5, no more than 6, no more than 7, no more than 8, no more than 9, no more than 10, no more than 11, no more than 12, no more than 13, no more than 14, no more than 15, no more than 16, no more than 17, no more than 18, no more than 19, or no more than 20) amino acid variation relative to amino acids 21-552 of SEQ ID NO: 12, where the resulting sequence is not found in a naturally occurring protein, e.g., a wild-type protein.
  • the amino acid sequence has altered glycosylation patterns compared to a naturally occurring protein, including but not limited to, an amino acid sequence produced in non-human cells, e.g., yeast cells, insect cells, CHO cells, etc.
  • the present invention provides for an isolated nucleic acid molecule, which is a cDNA molecule, comprises nucleic acids 405-2063 of the nucleic acid sequence set forth in SEQ ID NO: 11 as shown in FIG. 6D or a sequence that is at least 80%, at least 85%, at least 90%, or at least 95% homologous thereto, including sequences that are not identical to nucleic acids 405-2063 of SEQ ID NO: 11.
  • the present invention provides for an isolated nucleic acid molecule, which is a cDNA molecule, comprises nucleic acids 411-2063 of the nucleic acid sequence set forth in SEQ ID NO: 11 as shown in FIG. 6D or a sequence that is at least 80%, at least 85%, at least 90%, or at least 95% homologous thereto, including sequences that are not identical to nucleic acids 411-2063 of SEQ ID NO: 11.
  • the present invention provides for an isolated nucleic acid molecule, which is a cDNA molecule, comprises nucleic acids 465-2063 of the nucleic acid sequence set forth in SEQ ID NO: 11 as shown in FIG. 6D or a sequence that is at least 80%, at least 85%, at least 90%, or at least 95% homologous thereto, including sequences that are not identical to nucleic acids 465-2063 of SEQ ID NO: 11.
  • said nucleic acid molecule is operably linked to a promoter, which may be a heterologous promoter.
  • the nucleic acid molecule comprises a cDNA molecule.
  • the isolated nucleic acid molecule is comprised in a vector which may, for example, be an expression vector or a virus.
  • the present invention provides for a host cell comprising a vector that comprises the nucleic acid molecules described herein.
  • Sequence homology or sequence identity may be measured using sequence analysis software (for example, Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705, BLAST, BESTFIT, GAP, or PILEUP/PRETTYBOX programs). Such software matches identical or similar sequences by assigning degrees of homology to various substitutions, deletions, and/or other modifications.
  • sequence analysis software for example, Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705, BLAST, BESTFIT, GAP, or PILEUP/PRETTYBOX programs.
  • Such software matches identical or similar sequences by assigning degrees of homology to various substitutions, deletions, and/or other modifications.
  • a BLAST program may be used, with a probability score between e-3 and e-100 indicating a closely related sequence.
  • the present invention provides for an isolated polypeptide encoded by any of the nucleic acid molecules described herein, including cDNA molecules.
  • the present invention provides for an isolated polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 12 as shown in FIG. 6E , but lacking the amino acid sequence set forth in SEQ ID NO: 3 (provided below) or a sequence that is at least 80%, at least 85%, at least 90%, or at least about 95% homologous thereto, including sequences that are not identical to SEQ ID NO: 12.
  • the present invention provides for an isolated polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 12 as shown in FIG. 6E , but lacking the amino acid sequence set forth in SEQ ID NO: 4 (provided below) or a sequence that is at least 80%, at least 85%, at least 90%, or at least about 95% homologous thereto, including sequences that are not identical to SEQ ID NO: 12.
  • the polypeptide comprises amino acids 3 to 552 of SEQ ID NO: 12. In certain non-limiting embodiments, the polypeptide comprises amino acids 21 to 552 of SEQ ID NO: 12.
  • the isolated polypeptide comprises an amino acid sequence that comprises at least one (e.g., no more than 2, no more than 3, no more than 4, no more than 5, no more than 6, no more than 7, no more than 8, no more than 9, no more than 10, no more than 11, no more than 12, no more than 13, no more than 14, no more than 15, no more than 16, no more than 17, no more than 18, no more than 19, or no more than 20) amino acid variation relative to SEQ ID NO: 12, where the resulting sequence is not found in a naturally occurring protein, e.g., a wild-type protein.
  • the amino acid sequence comprises at least one amino acid variation relative to SEQ ID NO: 12 but lacks the amino acid sequence set forth in SEQ ID NO: 3.
  • the amino acid sequence comprises at least one amino acid variation relative to SEQ ID NO: 12 but lacks the amino acid sequence set forth in SEQ ID NO: 4.
  • the amino acid sequence has altered glycosylation patterns compared to a naturally occurring protein, including but not limited to, an amino acid sequence produced in non-human cells, e.g., yeast cells, insect cells, CHO cells, etc.
  • Amino acid variations or modifications may include amino acid substitutions, additions and/or deletions. Variations or modifications may be introduced by standard techniques known in the art, such as site-directed mutagenesis and PCR-mediated mutagenesis.
  • the at least one amino acid variation comprises at least one amino acid conservative modification.
  • Amino acids can be classified into groups according to their physicochemical properties such as charge and polarity. Conservative amino acid substitutions are ones in which the amino acid residue is replaced with an amino acid within the same group.
  • amino acids can be classified by charge: positively-charged amino acids include lysine, arginine, histidine, negatively-charged amino acids include aspartic acid, glutamic acid, neutral charge amino acids include alanine, asparagine, cysteine, glutamine, glycine, isoleucine, leucine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine.
  • positively-charged amino acids include lysine, arginine, histidine
  • negatively-charged amino acids include aspartic acid
  • glutamic acid neutral charge amino acids include alanine, asparagine, cysteine, glutamine, glycine, isoleucine, leucine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine.
  • amino acids can be classified by polarity: polar amino acids include arginine (basic polar), asparagine, aspartic acid (acidic polar), glutamic acid (acidic polar), glutamine, histidine (basic polar), lysine (basic polar), serine, threonine, and tyrosine; non-polar amino acids include alanine, cysteine, glycine, isoleucine, leucine, methionine, phenylalanine, proline, tryptophan, and valine.
  • the polypeptide comprises amino acids 3 to 552 of SEQ ID NO: 12. In certain non-limiting embodiments, the polypeptide comprises amino acids 21 to 552 of SEQ ID NO: 12.
  • the present invention provides for an isolated polypeptide comprising the amino acid sequence set forth in residues 1-552 of SEQ ID NO: 12 as shown in FIG. 6E .
  • the polypeptide has an estimated molecular weight of about 61 kDa (e.g., 61.08 kDa).
  • the present invention provides for an isolated polypeptide comprising the amino acid sequence set forth in residues 3-552 of SEQ ID NO: 12 as shown in FIG. 6E .
  • the polypeptide has an estimated molecular weight of about 61 kDa (e.g., 60.82 kDa).
  • the present invention provides for an isolated polypeptide comprising the amino acid sequence set forth in residues 21-552 of SEQ ID NO: 12 as shown in FIG. 6E .
  • the polypeptide has an estimated molecular weight of about 59 kDa (e.g., 58.71 kDa).
  • the polypeptide has an estimated molecular weight of between 58-64, e.g., about 61 kDa, or about 59 kDa.
  • the polypeptide is linked to a heterologous peptide tag of between about 2-20 amino acids, or between about 3-15 amino acids.
  • the polypeptide is detectably labeled.
  • the polypeptide contains at least one conservative amino acid substitution relative to the wild-type sequence.
  • the present invention provides for an antibody that binds to one of the above-described polypeptides, but that does not bind to a wild-type ALK polyeptide, e.g., one comprising the amino acid sequence set forth in SEQ ID NO: 1.
  • TALKs may be performed by any method known in the art.
  • a TALK may be detected in the form of a mRNA transcript, a corresponding cDNA molecule, its encoded polypeptide, or an ATI site in intron 19 of ALK.
  • the TALK is ALK ATI .
  • a mRNA transcript encoding a TALK may be detected by a method comprising probe hybridization (e.g., probe-based NanoString nCounter assay), amplification, reverse transcription, polymerase chain reaction (PCR, e.g., reverse transcription polymerase chain reaction (RT-PCR), quantitative reverse transcriptase PCR, real-time PCR, quantitative real-time PCR), Northern blot, sequencing, microarray, or a combination thereof.
  • probe hybridization e.g., probe-based NanoString nCounter assay
  • PCR polymerase chain reaction
  • RT-PCR reverse transcription polymerase chain reaction
  • quantitative reverse transcriptase PCR e.g., reverse transcriptase PCR
  • real-time PCR e.g., quantitative reverse transcriptase PCR
  • quantitative real-time PCR e.g., Northern blot, sequencing, microarray, or a combination thereof.
  • a TALK polypeptide may be detected by a method comprising antibody binding, immunohistochemistry, Western blot, functional (e.g., kinase) assay, enzyme linked immunosorbent assays (ELISAs), radioimmunoassays (RIA), enzyme immunoassays (EIA), mass spectrometry, 1-D or 2-D gel-based analysis systems (e.g., Polyacrylamide gel electrophoresis (PAGE)), immunoprecipitation, or a combination thereof.
  • functional e.g., kinase
  • ELISAs enzyme linked immunosorbent assays
  • RIA radioimmunoassays
  • EIA enzyme immunoassays
  • mass spectrometry e.g., 1-D or 2-D gel-based analysis systems (e.g., Polyacrylamide gel electrophoresis (PAGE)), immunoprecipitation, or a combination thereof.
  • PAGE Polyacrylamide gel electrophoresis
  • a TALK cDNA molecule may be detected by PCR.
  • an ATI site in intron 19 of an ALK gene may be detected by a method comprising Northern blot, Chromatin immunoprecipitation (ChIP)-seq, ChIP-qPCR, Rapid Amplification of cDNA Ends (RACE)-PCR, or a combination thereof.
  • a method comprising Northern blot, Chromatin immunoprecipitation (ChIP)-seq, ChIP-qPCR, Rapid Amplification of cDNA Ends (RACE)-PCR, or a combination thereof.
  • a detection method may be practiced in a cell or tissue or cell lysate collected from a subject.
  • the cell may be collected as a sample, for example a sample of a tumor or neoplasm or other lesion.
  • the sample may be a peripheral blood sample or other fluid sample (e.g., pleural effusion).
  • the transcriptional start site and 5′-end of a TALK transcript may be ascertained using the 5′-RACE technique.
  • a tobacco-acid-pyrophosphatase 5′-RACE technique may be performed according to the manufactures protocol (ExactSTART Eukaryotic mRNA RACE Kit, #ES80910, Epicentre) using the following primers: 5′-TCATACACATACGATTTAGGTGACACTATAGAGCGGCCGCCTGCAGGAAA-3′ [SEQ ID NO: 5]; reverse 5′-CAGGTCACTGATGGAGGAGGTCTTGCCAGCAAAGCA-3′ [SEQ ID NO: 6].
  • the 5′-RACE products may then be sequenced, for example using an Illumina MiSeq System with a 150 bp paired-end protocol according to the manufactures protocol.
  • the following reverse primers may be used: forward primer 5′-CTAATACGACTCACTATAGGGC-3′ [SEQ ID NO: 7], reverse primer 5′-ACACCTGGCCTTCATACACCTCC-3′ [SEQ ID NO: 8].
  • a cDNA molecule of a TALK (e.g., the ALK ATI ) transcript may be amplified using the ALK specific primers 5′-CACCATCCCATCTCCAGTCTGCTTC-3′ [SEQ ID NO: 9] and 5′-AGAGAAGTGAGTGTGCGACC-3′ [SEQ ID NO: 10].
  • the presence of a TALK may be detected using an ALK-specific antibody as well as a technique that demonstrates a molecular weight lower than wild-type protein, such as but not limited to polyacrylamide gel electrophoresis and Western blot.
  • Non-limiting examples of antibodies that may be used include Anti- ⁇ -Tubulin antibody (#T9026-.5ML), Anti-V5 (#MA1-81617), and anti-HA3F10 (#12158167001), Phospho-ALK (Tyr1278/1282/1283) Rabbit mAb (#3983), Phospho-ALK (Tyr1604) Rabbit mAb (#3341), ALK (D5F3) Rabbit mAb (#3633), Phospho-Akt (Ser473) (#4060), Akt (#4685), Phospho-Stat3 (Tyr705) (#9145), Stat3 (#4904), Phospho-S6 (Ser235/236) (#4858), S6 (#2217), Phospho-p44/42 MAPK (Erk1/2) (Thr202/Tyr204) (#4370), p44/42 MAPK (Erk1/2) (#4695), Phospho-MEK1/2 (Ser221) (
  • a TALK may be detected via the presence of an ATI site in intron 19 of ALK, for example using ChIP-seq and ChIP-qPCR.
  • the chromatin marks ‘trimethylated histone H3 lysine 4 (H3K4me3)’ and ‘RNApol-II’ are well characterized to enrich at poised and actively transcribed ATI site. 17-19
  • NanoString nCounter assay with probe sets in ALK exons 1-19, exons 20-29 and intron 19 may be used (see, for FIG. 1H and its legend) to distinguish wild-type ALK (ALK WT ), translocated ALK and ALK ATI transcripts.
  • NanoString nCounter assay is based on digital detection and direct molecular barcoding of target molecules through the use of a color coded probe pair.
  • the probe pair consists of a Reporter Probe, which carries the signal on its 5′ end, and a Capture Probe which carries a biotin on the 3′ end. 51
  • a TALK may be detected via its cellular localization, in that TALK has a greater presence in the nucleus relative to wild-type ALK.
  • Such studies may be performed, for example, using immunohistochemistry and primary or secondary fluorescent antibody probes.
  • the present invention provides for a method of diagnosing a cell as a cancer cell comprising detecting the presence of a TALK in a cell, where the presence of a TALK indicates that the cell is a cancer cell.
  • the cell may be in a sample collected from a subject.
  • a sample includes, but is not limited to, a clinical sample, cells in culture, cell supernatants, cell lysates, serum, blood plasma, biological fluid (e.g., lymphatic fluid) and tissue samples.
  • the source of the sample may be solid tissue (e.g., from a fresh, frozen, and/or preserved organ, tissue sample, biopsy or aspirate), blood or any blood constituents, bodily fluids (such as, e.g., urine, lymph, cerebral spinal fluid, amniotic fluid, peritoneal fluid or interstitial fluid), or cells from the individual, including circulating tumor cells.
  • solid tissue e.g., from a fresh, frozen, and/or preserved organ, tissue sample, biopsy or aspirate
  • blood or any blood constituents e.g., blood or any blood constituents, bodily fluids (such as, e.g., urine, lymph, cerebral spinal fluid, amniotic fluid, peritoneal fluid or interstitial fluid), or cells from the individual, including circulating tumor cells.
  • bodily fluids such as, e.g., urine, lymph, cerebral spinal fluid, amniotic fluid, peritoneal fluid or interstitial fluid
  • cells from the individual including circulating tumor cells.
  • the cell or another cell from the same subject may be tested for other markers indicative of a diagnosis of cancer, as are known in the art.
  • Non-limiting examples of cancers which may be indicated by the presence of a TALK include melanoma, thyroid carcinoma (e.g., anaplastic thyroid carcinoma), lung adenocarcinoma, lung squamous cell carcinoma, renal clear cell carcinoma, and breast cancer.
  • the cancer is melanoma.
  • the present invention provides a method of determining whether an anti-cancer effect is likely to be produced in a cancer by an ALK inhibitor, comprising determining whether one or more cell of the cancer contains a detectable TALK, wherein the presence of a detectable TALK in the cell indicates that an ALK inhibitor would have an anti-cancer effect on the cancer.
  • the present invention provides for a method of determining the likelihood that a subject having a cancer may obtain therapeutic benefit from therapy with an ALK inhibitor, comprising determining whether a cancer cell of the subject contains a detectable TALK, where the presence of a detectable TALK indicates that the subject is more likely to benefit from ALK inhibitor therapy than a subject who lacks TALK or an activating genetic mutation of ALK.
  • the presence of a detectable TALK comprises the presence of a detectable TALK mRNA transcript, a detectable TALK cDNA molecule corresponding thereto, a detectable TALK polypeptide encoded thereby, and/or a detectable ATI site in intron 19 of an ALK gene in the cell.
  • a cancer cell of a subject may have a wild-type (or normal) ALK locus and yet express a TALK, and therefore would not be identified as having an activated ALK by genomic testing.
  • the invention provides for a method of treating a subject suffering from a cancer, comprising (i) determining the likelihood that the subject would obtain therapeutic benefit from therapy with an ALK inhibitor, comprising determining whether a cancer cell of the subject contains a detectable TALK, where the presence of a detectable TALK indicates that the subject is more likely to benefit from ALK inhibitor therapy than a subject who lacks TALK or an activating genetic mutation of ALK; (ii) where the subject is determined to be more likely to benefit from ALK inhibitor therapy, treating the subject with an ALK inhibitor, and (iii) where the subject is determined to not be more likely to benefit from ALK inhibitor therapy, treating the subject with an alternative therapy which is not an ALK inhibitor but which may be surgical treatment, radiation therapy, or chemotherapy.
  • Non-limiting examples of cancers which may be treated by the above method include melanoma, thyroid carcinoma (e.g., anaplastic thyroid carcinoma), lung adenocarcinoma, lung squamous cell carcinoma, renal clear cell carcinoma, thyroid cancer, and breast cancer.
  • the cancer is melanoma.
  • Non-limiting examples of ALK inhibitors which may be used in the above method include crizotinib (“Xalkori®”; (Pfizer); ceritinib (Zykadia®; also known as “LDK-378; Novartis), NVP-TAE684 (Novartis), alectinib (Chugai); AP26113 (Ariad); ASP-3026 (Astellas); CEP-37440 (Teva); NMS-E628 (Nerviano); PF-06463922 (Pfizer); TSR-011 (Tesoro); RXDX-101 (Ignyta Inc.), and X-396 (Xcovery).
  • the ALK inhibitor is crizotinib.
  • Subjects treated according to the invention with one or more ALK inhibitor may further be treated with one or more additional cancer treatment, including but not limited to, one or more BRAF inhibitor, one or more MEK inhibitor, one or more immunologic inhibitor (e.g., an anti-CTLA4 antibody, an anti-PD1 antibody, an anti-PD-L1 antibody), one or more CDK4 inhibitor, one or more CDK6 inhibitor, one or more alklyating agent, one or more topoisomerase inhibitor, one or more anti-metabolite, one or more anti-microtubule agent, one or more cytotoxic antibiotic, radiation therapy, chemotherapy, or a combination thereof.
  • one or more BRAF inhibitor e.g., an anti-CTLA4 antibody, an anti-PD1 antibody, an anti-PD-L1 antibody
  • CDK4 inhibitor e.g., an anti-CTLA4 antibody, an anti-PD1 antibody, an anti-PD-L1 antibody
  • CDK4 inhibitor e.g., a CDK4 inhibitor
  • CDK6 inhibitor e
  • the subject receives a combination of an anti-CTLA-4 antibody and an PD-1 antibody in addition to an ALK inhibitor.
  • the subject has been treated with a combination of an anti-CTLA-4 antibody and an anti-PD-1 antibody prior to the treatment with an ALK inhibitor.
  • the subject further receives radiation and/or chemotherapy.
  • the present invention provides for a kit comprising a means for detecting a TALK as described above, optionally together with written disclosure of one or more method set forth above, and optionally together with a positive control molecule such as a TALK protein and/or nucleic acid.
  • the kit comprises means for determining the level of a TALK mRNA transcript, a TALK cDNA molecule corresponding thereto, and/or a TALK polypeptide encoded thereby.
  • the means for determining the level of a TALK mRNA transcript may be probe hybridization (e.g., probe-based NanoString nCounter assay), polymerase chain reaction (PCR, e.g., RT-PCR, quantitative reverse transcriptase PCR, real-time PCR, or quantitative real-time PCR), Northern blot, sequencing, microarray, or a combination thereof.
  • the means for determining the level of a TALK cDNA molecule may be PCR.
  • the means for determining the level of a TALK polypeptide may be antibody binding, immunohistochemistry, Western blot, a functional (e.g., kinase) assay, enzyme linked immunosorbent assays (ELISAs), radioimmunoassays (RIA), enzyme immunoassays (EIA), mass spectrometry, 1-D or 2-D gel-based analysis systems (e.g., Polyacrylamide gel electrophoresis (PAGE)), immunoprecipitation, or a combination thereof.
  • a functional e.g., kinase
  • ELISAs enzyme linked immunosorbent assays
  • RIA radioimmunoassays
  • EIA enzyme immunoassays
  • mass spectrometry e.g., 1-D or 2-D gel-based analysis systems (e.g., Polyacrylamide gel electrophoresis (PAGE)), immunoprecipitation, or a combination thereof.
  • PAGE Polyacrylamide gel electrophores
  • the kit comprises means for determining the presence of an ATI site in intron 19 of an ALK gene in one or more cell of the cancer, which may be Northern blot, Chromatin immunoprecipitation (ChIP)-seq, ChIP-qPCR, Rapid Amplification of cDNA Ends (RACE)-PCR, or a combination thereof.
  • means for determining the presence of an ATI site in intron 19 of an ALK gene in one or more cell of the cancer which may be Northern blot, Chromatin immunoprecipitation (ChIP)-seq, ChIP-qPCR, Rapid Amplification of cDNA Ends (RACE)-PCR, or a combination thereof.
  • the present invention further provides for a means for determining the presence of a detectable TALK for use in a method of determining whether an anti-cancer effect is likely to be produced in a cancer by an ALK inhibitor, the method characterized by determining whether one or more cell of the cancer contains a detectable TALK, where the presence of a detectable TALK in the cell indicates that an ALK inhibitor would have an anti-cancer effect on the cancer.
  • kit may therefore comprise one or more nucleic acid probe (e.g., that hybridizes with a TALK mRNA transcript (including a color coded probe pair used in a NanoString nCounter assay), or that hybridizes with a TALK cDNA molecule), primer, and/or primer pair (e.g., that hybridizes with a TALK mRNA transcript or a TALK cDNA molecule), and/or one or more antibody, antibody fragment, or single chain antibody (e.g., that binds specifically to a TALK polypeptide), any of which are optionally directly labeled with a chemical, fluorescent, enzymatic or radioactive marker.
  • detection may be achieved via a secondary probe, enzyme, substrate, ligand, antibody, antibody fragment, single chain antibody, etc., using techniques known in the art.
  • the kit comprises the primer pair: 5′-TCATACACATACGATTTAGGTGACACTATAGAGCGGCCGCCTGCAGGAAA-3′ [SEQ ID NO: 5]; reverse 5′-CAGGTCACTGATGGAGGAGGTCTTGCCAGCAAAGCA-3′ [SEQ ID NO: 6], for example, but not by way of limitation, for use in a 5′-RACE technique.
  • the kit comprises the primer pair: forward primer 5′-CTAATACGACTCACTATAGGGC-3′ [SEQ ID NO: 7], reverse primer 5′-ACACCTGGCCTTCATACACCTCC-3′ [SEQ ID NO: 8], for example, but not by way of limitation, for use in a 5′-RACE technique.
  • the kit comprises the primer pair: 5′-CACCATCCCATCTCCAGTCTGCTTC-3′ [SEQ ID NO: 9] and 5′-AGAGAAGTGAGTGTGCGACC-3′ [SEQ ID NO: 10].
  • the kit comprises an antibody or a fragment thereof that binds to ALK in a region comprised in exons 20-29 and optionally an antibody or a fragment thereof that binds to ALK in a region comprised in exons 1-19.
  • the kit comprises means to identify an ATI site in intron 19 of ALK.
  • the kit comprises means to detect nuclear localization of TALK, for example a nucleic acid or antibody probe and a second nucleic acid or antibody probe that specifically binds to a binding partner in the nucleus or a chemical means of identifying or staining the nucleus.
  • Sequencing data were mapped to the human reference genome (hg19) using Bowtie or BWA and analyzed using publicly available software packages: SAMtools 26 , Tophat 27 , FusionSeq 28 , GATK 29 , Picard (http://picard.sourceforge.net) and IGV 30
  • RTK receptor tyrosine kinase genes
  • RNA-seq data was downloaded from the Broad Institute GTEx Genotype-Tissue Expression Portal (http://www.broadinstitute.org/gtex/) 2013_09_23 run using exon_quantification data from illuminahiseqrnaseqv2 unc_udu.
  • ALK ATI candidates were identified as samples with an ALK expression level of RSEM ⁇ 100, ⁇ 500 total reads across all ALK exons, and ⁇ 10 ⁇ greater average expression (by exon-level RPKM) in exons 20-29 compared to exons 1-19. To confirm ALK ATI expression, candidates were manually examined in IGV 30 .
  • a tobacco-acid-pyrophosphatase 5′-RACE technique according to the manufacture's protocol (ExactSTART Eukaryotic mRNA RACE Kit, #ES80910, Epicentre) using the following primers: 5′-TCATACACATACGATTTAGGTGACACTATAGAGCGGCCGCCTGCAGGAAA-3′ [SEQ ID NO: 5]; reverse 5′-CAGGTCACTGATGGAGGAGGTCTTGCCAGCAAAGCA-3′ [SEQ ID NO: 6].
  • the 5′-RACE products were sequenced on an Illumina MiSeq System with a 150 bp paired-end protocol according to the manufacture's protocol.
  • the sequencing reads were mapped to the human reference genome (hg19) using BWA, analyzed using Tophat 27 and visualized using IGV 30 .
  • ALK intron 19 The continuous transcription starting in ALK intron 19 was confirmed with an independent oligonucleotide-based 5′-RACE kit (SMARTerTM RACE cDNA Amplification Kit, #634923, Clontech) according to the manufacture's protocol using the following reverse primers: forward primer 5′-CTAATACGACTCACTATAGGGC-3′ [SEQ ID NO: 7], reverse primer 5′-ACACCTGGCCTTCATACACCTCC-3′ [SEQ ID NO: 8].
  • the RACE cDNA products were cloned into plasmids (Zero Blunt® TOPO® PCR Cloning Kit, #K2800-20, Invitrogen) and were analyzed with Sanger sequencing using standard procedures.
  • Two lung cancer cell lines (H3122, H2228) with EML4-ALK translocations were used as controls, and as expected, both controls showed the EML4 gene next to ALK exon 20.
  • ChIP Chromatin Immunoprecipitation
  • Chromatin was isolated from human tumor tissue and cell lines. Fresh-frozen human tumor tissue (MM-15, MM-74, ATC-28) was sectioned with a microtome and cross-linked in 1% paraformaldehyde for 15 min. The cross-linked tissue samples were quenched in 125 mM Glycine for 10 min, washed in PBS, re-suspended in lysis buffer, and dounced in a Tenbroeck-style tissue grinder and conicated. 32 Chromatin isolation from the cell-lines H3122 and SKMEL-524 cells and immunoprecipitation was performed as previously described 32 .
  • Solubilized chromatin from human tumors and cell lines was immune-precipitated with antibodies against H3K4me3 (#39159, Active Motif), H3K27ac (#Ab4729, Abcam), and RNA-pol II (#39097, Active Motif).
  • ChIP-seq was performed on an Illumina HiSeq2500 with 51-bp single reads. Reads were aligned to the human genome hg19 using the Bowtie alignment software within the Illumina Analysis Pipeline. Duplicate reads were eliminated for subsequent analysis. Peak calling was performed using MACS 1.4 comparing immune-precipitated chromatin with input chromatin. 33 ChIP-qPCR was performed on a ViiATM 7 Real Time PCR System (Life Technologies) using Power SYBR Master Mix (#4367659, Life Technology). ChIP-qPCR primers were designed using primer blast (http://www.ncbi.nlm.nih.gov/tools/primer-blast) and are described in FIGS. 7A and 7B .
  • Targeted sequencing of the entire ALK locus was performed using custom hybridization capture probes tiling hg19 chr2:29400000-30300000 (Roche/NimbleGen's SeqCap EZ). This region encompassed the entire genomic footprint of ALK as well as ⁇ 150 kb of upstream sequence. After the genomic DNA was fragmented (E220, Covaris), barcoded sequence libraries (New England Biolabs, Kapa Biosystems) were prepared, and hybridization capture on barcoded pools was performed using custom probes (Nimblegen SeqCap).
  • genomic DNA 250 ng was used for library construction from 7 separate samples: 2 melanoma tumors (MM-15, MM-74), 1 anaplastic thyroid carcinoma (ATC-28), 2 lung cancer cell lines (H3122, H2228) with EML4-ALK translocations, 1 melanoma (SKMEL-28) and 1 control pool of 10 “normal” blood samples.
  • Libraries were pooled at equimolar concentrations (100 ng per library) and used in the capture reaction as previously described.
  • 34 To prevent off-target hybridization, we spiked in a pool of blocker oligonucleotides complementary to the full sequences of all barcoded adaptors. The captured libraries were sequenced on an Illumina HiSeq 2500 to generate 75-bp paired-end reads. Sequence data were de-multiplexed using CASAVA, and aligned to the reference human genome (hg19) using BWA. 35
  • Custom capture of the entire ALK locus was performed using custom hybridisation capture probes tiling the entire genomic footprint of ALK (900 kb, chr2:29400000-30300000) followed by bisulfite sequencing. After fragmentation (E220, Covaris) of 3 g genomic DNA of each sample (MM-15, ATC-28, H3122 and SKMEL-28), libraries were prepared with the KAPA Hyper Prep Kit (#KR0961, Kapa Biosystems) without PCR amplification to preserve the methylation status. 1 ⁇ g of each barcoded library was pooled at equimolar concentrations and captured according to the manufacturer's protocol (Roche/NimbleGen's SeqCap EZ).
  • the non-biotinylated tumor/cell line DNA was dissociated from the biotinylated capture beads with 0.5M NaOH.
  • the single stranded eluted DNA was used for bisulfite conversion using the EZ DNA Methylation-GoldTM Kit (#D5005, Zymo Research) according to the manufacturer's protocol, except for the 98° C. denaturation step.
  • the KAPA HiFi Uracil PCR polymerase (#KK280, Kapa Biosystems) was used to amplify the library, the reaction with Agencourt AMPure XP beads (A63881, Beckman Coulter) was purified, and the library was sequenced on an Illumina MiSeq with a 150 bp paired-end protocol according to the manufacturer's instructions. Sequence data were aligned to hg19 and analyzed using BISMARK 53 . The methylation level at CpG sites was compared across all samples; no methylation was detected in the CHG and CHH contexts. Methylation was first computed as the number of methylated CpG reads vs.
  • Genomic DNA samples were labeled using a Bioprime Array CGH Genomic Labeling Kit (#18095-011, Life Technologies) according to the manufacturer's instructions. Briefly, 1 ⁇ g tumor DNA and reference DNA (#G1471, Promega) were differentially labeled with dCTP-Cy5 (#45-001-291, GE Healthcare), and dCTP-Cy3 (#45-001-290, GE Healthcare). Genome-wide analysis of DNA copy number changes was conducted using an oligonucleotide SurePrint G3 Human CGH Microarray (#G4447A, Agilent) containing 1 million probes according to the manufacturer's protocol. Slides were scanned using Agilent's microarray scanner G2505B and analyzed using Agilent Genomic Workbench.
  • FISH Interphase Fluorescence In situ Hybridization
  • a commercially available ALK break-apart probe (#06N38-020, Abbott) was used according to the manufacturer's protocol. The probes were hybridized on 5 ⁇ m-thick tissue sections. The number and localization of the hybridization signals was assessed in a minimum of 100 interphase nuclei with well-delineated contours. At least 10% of neoplastic cells had to show a split signal to report an ALK rearrangement.
  • nCounter Analysis System (NanoString Technologies) were reported previously. 39 In brief, two sequence-specific probes were constructed for ALK exons 1-19, intron 19, and exons 20-29, respectively. Four control genes (RPS13, RPL27, RPS20, ACTB) were used for normalization. The probes were complementary to a 100 base pair region of the target mRNA and are listed in FIG. 1H . 100 ng of total RNA from each sample was hybridized, the raw data were normalized to the standard curve generated via the nCounter system, and the average value of the two probes in each target region (exons 1-19, intron 19, exons 20-29) were printed in bar charts using GraphPad Prism software 6.0.
  • NIH-3T3 mouse embryonic fibroblast cells were obtained from the ‘American Type Culture Collection’ (#CRL-1658, ATCC) and were maintained in DMEM.
  • the Interleukin 3 (IL-3) dependent murine pro B cell line, Ba/F3 was obtained from ‘The Deutsche Sammlung von Mikroorganismen und Zellkulturen’ (#ACC-300, DSMZ) and was cultured in RPMI supplemented with Interleukin 3 (1 ng ml; #403-ML-010, R&D Systems).
  • Melan-a cells were provided by Dr. Dorothy Bennett (St.
  • ALK ATI RNA from MM-15 was reverse-transcribed with anchored oligo(dT) primers into cDNA (#04379012001, Roche), PCR amplified with ALK specific primers 5′-CACCATCCCATCTCCAGTCTGCTTC-3′ [SEQ ID NO: 9] and 5′-AGAGAAGTGAGTGTGCGACC-3′ [SEQ ID NO: 10], and the PCR product was cloned into a pENTR vector (#K2400-20, Life Technologies).
  • ALK plasmid The full-length ALK plasmid (HsCD00079531) was purchased from the DF/HCC DNA Resource Core (http://plasmid.med.harvard.edu), and EML4-ALKv1 was synthesized at GeneArt (Life Technologies). Site-directed mutagenesis was performed using QuikChange (#200523, Agilent): For kinase-dead ALK ATI (ALK ATI-KD ), the lysine in the ATP-binding site of the kinase domain was mutated to methionine (p.K1 150M) in ALK ATI , and for ALK F1174L , a p.F1174L mutation was introduced into ALK WT .
  • ALK ATI ALK ATI-KD
  • Plasmids were sub-cloned into a pMIG-w vector 41 (#12282, www.addgene.org), resulting in MSCV-ALK variant -IRES-GFP constructs, which were confirmed by digestion and sequencing.
  • An empty pMIG-w vector with green-fluorescent-protein (GFP) was used as control for all ALK expression experiments.
  • GFP green-fluorescent-protein
  • ALK ATL was cloned into pcDNA3.1/nV5-Dest (#12290-010, Life Technologies) and MSCV N-HA FLAG-Dest (#41033, Addgene).
  • a triple modality retroviral reporter plasmid red fluorescent protein (RFP)—thymidine kinase—luciferase) 42 was used.
  • Retrovirus were produced in 293T cells by standard methods using ecotropic or amphotropic packaging vector and XtremeGene 9 (#06365809001, Roche). We harvested the virus-containing supernatant were harvested for 48, 64, 64 h and 72 hours after transfection. The supernatant was pooled, filtered through a 0.45 ⁇ m PVDF membrane, and used for transduction in the presence of polybrene (8 ⁇ g ml-1). Transduced stably expressing eGFP+ or RFP+ cells were sorted with a FACSAria II (BD Biosciences).
  • V5-ALK ATI and HA-ALK ATI was transiently transfected into 293T cells and after 24 hours, cells were lysed in 10 mM Tris HCl pH 7.5, 1% Triton X-100, 150 mM NaCl, 1 mM EDTA, 1 mM DTT, 1 mM PMSF, proteinase/phosphatase inhibitors.
  • Stably transduced NIH-3T3 cells were grown in a 15 cm dish, washed in PBS, and lysed in 20 mM Tris pH 8.0, 1% NP-40, 125 mM NaCl, 2.5 mM MgCl2, 1 mM EDTA with proteinase/phosphatase inhibitor. Lysates were incubated on ice, centrifuged, pre-cleared with 25 ⁇ l Protein A/G UltraLink Resin (#53133, Thermo Scientific) for 30 min at 4° C. under rotation, and immunoprecipitated with 10 ⁇ l ALK (D5F3) XP® Rabbit mAb (#3633) and 25 ⁇ l Protein A/G UltraLink Resin.
  • the immunoprecipitated material was washed and used according to the instructions of Universal Tyrosine Kinase Assay Kit (#MK410, Clontech). After the enzymatic reaction, the immunoprecipitated material was mixed with 4 ⁇ SDS loading buffer for immunoblotting.
  • Stably transduced NIH-3T3 cells were grown on coverslips, fixed in 4% formaldehyde, washed in PBS for 15 min at room temperature, washed in PBS for 10 min, and incubated in blocking solution (5% goat serum, 0.1% Triton X-100 in PBS) for 1 hour at room temperature. After aspirating blocking solution, cells were incubated with an ALK (D5F3) Rabbit mAb (#3633, Cell Signaling Technology) diluted 1:1000 in blocking buffer overnight at 4° C. After washing the cells 3 ⁇ with 0.05% Tween 20 and PBS, cells were incubate with a secondary antibody (#A-11012, Life Technologies) diluted 1:500 in blocking buffer for 2 hours at room temperature. After washing in PBS, slides were mounted with Prolong® Gold Antifade Reagent with DAPI (#8961, Cell Signaling Technology) and imaged with a Leica TCS SP5 II confocal microscope.
  • Cell lysates were prepared in RIPA buffer (#9806, Cell Signaling) supplemented with Halt protease and phosphatase inhibitor cocktail (#78440, Thermo Scientific). Equal amounts of protein, as measured by BCA protein assay (#23225, Thermo Scientific), were resolved in NuPAGE® Novex® 4-12% Bis-Tris Protein Gels (#NP0321BOX, Life Technologies) and transferred electrophoretically onto a Nitrocellulose 0.45 ⁇ m membrane (#162-0115, BioRad). Membranes were blocked for 1 hour at room temperature in 50% Odyssey Blocking Buffer in PBS (#927-40000, LI-COR) and were incubated overnight at 4° C.
  • the long terminal repeat in ALK intron 19 at the ATI site was amplified using genomic DNA from patient MM-15 and 5′-GTCCTCATGGCTCAGCTTGT-3′ and 5′-AGCACTACACAGGCCACTTC-3′ primers.
  • the PCR product (chr2:29,446,444-29,447,174; 731 bp) was cloned into pGL4.14-firefly luciferase vector (#E6691, Promega).
  • LTR16B2 10 5 cells were transfected with 500 g pGL4.14-LTR16B2 or vector alone; as internal control, 200 ⁇ g pRL-TKRenilla luciferase reporter vector (#E2241, Promega) was co-transfected. Luciferase activity was measured using Dual-Glo Luciferase Assay System (#E2920, Promega) 48 hours after transfection. Promoter activity was calculated by normalising firefly luciferase activity to the control Renilla luciferase activity and compared between pGL4.14-LTR16B2 and vector alone.
  • Flow cytometry analysis for in vitro transformation assays with Ba/F3 cells was performed on a LSRFortessa (BD Biosciences). GFP- or RFP-positive cells were sorted using a FITC (blue laser) or PE (yellow laser) channel, respectively, on a FACSAria II configured with 5 lasers (BD Biosciences).
  • FITC blue laser
  • PE yellow laser
  • Ba/F3 cells were stably transduced with MSCV-ALK variant -IRES-GFP constructs with a multiplicity of infection (MOI) of ⁇ 0.26. Based on the MOI calculations, ⁇ 78% of cells were uninfected, ⁇ 20% of cells were infected with one virus particle, and ⁇ 2% of cells were infected by more than one viral particle. Stably transduced Ba/F3 cells were cultured in RPMI medium supplemented with IL-3 (1 ng ml-1) and the transduction rate of ⁇ 20% was validated using flow cytometry for GFP, that was co-expressed with the ALK variants.
  • MOI multiplicity of infection
  • transduced Ba/F3 cells were transferred into IL-3 depleted RPMI medium and cell growth was quantified every 2-4 days with a luminescence assay (#G7571, Promega).
  • ALK inhibitor-dose response curve 2000 Ba/F3 cells were plated in quadruplicates in wells of 96-well plates with increasing concentrations of the ALK inhibitors crizotinib (#C-7900, LC laboratories), TAE684 (#CT-TAE684, ChemieTek), or ceritinib (#CT-LDK378, ChemieTek) as indicated. All drugs were suspended in DMSO.
  • the cell viability was assessed after 72 hours by a luminescence assay CellTiter-Glo (#G7571, Promega). Results were normalized to growth of cells in a medium containing an equivalent volume of DMSO. The inhibition curve was determined with GraphPad Prism 6.0 software using the ‘log(inhibitor) vs. response—variable slope’ non-linear regression model. For western blot analysis, 10 million Ba/F3 cells were harvested after 2 hours treatment with crizotinib, washed in ice-cold PBS and lysed in RIPA buffer (#9806, Cell Signaling).
  • mice were injected with the stably transduced NIH-3T3 cells expressing a luciferase reporter construct and the indicated plasmids. When the tumors reached an average size of 200-250 mm 3 , mice were randomized in a vehicle and or treat group. Mice were orally gavaged once a day with crizotinib (100 mg/kg/d) or vehicle. 8 tumors were measured with calipers every 2 to 3 days and growth curves were visualized with Prism GraphPad 6.0.
  • tumor growth was monitored by bioluminescence imaging of anesthetized mice by retro-orbitally injecting d-luciferin (150 mg per kg body weight) and imaging with the IVIS Spectrum Xenogen machine (Caliper Life Sciences). After euthanizing the mice, tumors were explanted either lysed in RIPA buffer (#9806, Cell Signaling Technology), or fixed overnight in 4% paraformaldehyde, washed, embedded in paraffin, and sectioned for hematoxylin-and-eosin (HE) staining or immunohistochemistry.
  • RIPA buffer #9806, Cell Signaling Technology
  • HE hematoxylin-and-eosin
  • transcriptome analyses of metastatic melanoma and thyroid carcinoma were performed.
  • An algorithm 9 was used to investigate the differential expression of exons in receptor tyrosine kinases (RTKs).
  • RTKs receptor tyrosine kinases
  • the analysis was focused on transcripts with a high expression of the kinase domain.
  • a novel ALK transcript in two melanoma (MM-15, MM-74) and in one anaplastic thyroid carcinoma (ATC-28) was identified.
  • the novel transcript contained the ALK exons 20-29 preceded by ⁇ 400 base pairs of intron 19, but not exons 1-19 ( FIGS. 1A and 5A ). This expression pattern was distinct from ALK wild-type (ALT WT ) and ALK translocations.
  • the ALT WT shows expression of all exons, but no expression of introns ( FIGS. 1A, 5B ), and is commonly found in neuroblastoma in association with activating mutations. 10-13 ALK translocations are observed in various cancer types, including lymphoma, sarcoma, and lung cancer and typically occur in intron 19. 14-16 Due to preserved splice sites, ALK translocations usually encompass ALK exons 20-29 with little intronic expression ( FIGS. 1A and 5C ). ALK WT overexpression 10, 58-60 and amplification 10,60 , activating ALK mutations 10,11-13 , and ALK translocations 15,16 are well-established oncogenic drivers in various cancer types, including melanocytic tumors 62 .
  • ALK ATI alternative transcriptional initiation
  • 5′-RACE 5′-rapid amplification of cDNA ends
  • the ATI site was mapped to a 25-bp region in intron 19 and the presence of the novel transcript was confirmed and termed as ALK ATI by Northern blot ( FIGS. 1B and 6A-6G ).
  • ChIPseq and ChIP-qPCR showed that only ALK ATI -expressing tumors, but not the controls, had significant enrichment of the chromatin marks trimethylated histone H3 lysine 4 (H3K4me3) and RNA polymerase II at the ATI site, which are characteristic for active promoters 17,18 ( FIGS. 1C-1E and 7A-7D ).
  • H3K4me3 trimethylated histone H3 lysine 4
  • RNA polymerase II RNA polymerase II
  • ALK ATI was expressed in ⁇ 11% of melanoma (38 of 334 tumors) and sporadically in other cancer types, including lung adenocarcinoma, lung squamous cell carcinoma, clear cell renal cell carcinoma, and breast carcinoma ( FIGS. 8A-8E ).
  • No ALK ATI expression was found in more than 1600 samples from 43 different normal tissues in the Genotype-Tissue Expression (GTEx) RNA-seq dataset 1 , indicating that ALK ATI is primarily expressed in cancers, particularly melanoma.
  • GTEx Genotype-Tissue Expression
  • FIG. 1H a NanoString nCounter assay with probe sets in ALK exons 1-19, intron 19, and exons 20-29 was developed 51 ( FIG. 1H ). This assay was able to distinguish ALK ATI , ALK WT and translocated ALK and identified additional ALK ATI -expressing tumors derived from both fresh-frozen and formalin-fixed, paraffin-embedded (FFPE) clinical specimens ( FIG. 1F ).
  • FFPE paraffin-embedded
  • FIGS. 9C and 9D 16 genome-wide array-CGH was used, but no genetic alterations was discovered ( FIGS. 9C and 9D ).
  • ALK ATI could arise through genetic alterations that create a de novo ATI site similar to the recently described TERT promoter mutations in melanoma.
  • 23,24 ultra-deep targeted sequencing of the entire ALK locus was performed. No recurrent single nucleotide variations (SNVs), insertions, or deletions were identified in the ALK exons and introns ( FIGS. 10A-10D ). In summary, no genomic aberrations that could account for the de novo expression of ALK ATI were found.
  • the ALK ATI region contains transposable elements, including a long-terminal repeat (LTR) in intron 19 and a long-interspaced element (LINE) in intron 18, both of which can regulate transcription 44,45 ( FIG. 11A ).
  • LTR long-terminal repeat
  • LINE long-interspaced element
  • H3K27ac enrichment (a histone mark characteristic of active promoters and enhancers 47 ) was found in all analyzed melanoma samples, but not in the control lung cancer cell lines or in the 17 non-melanoma cell lines in ENCODE ( FIGS. 11E and 11F ).
  • the proximal cis-regulatory region was defined as chr2:29,445,000-29,447,100 and the potential transcription factor binding motifs were bioinformatically determined. 48 ( FIG. 11H ).
  • ALK ATI transcript 5′-RACE (5′-rapid amplification of cDNA ends) coupled with next-generation sequencing was used to determine the ALK ATI transcript.
  • the ATI site was mapped to a 25 base pairs (bps) region in intron 19, approximately 400 bp upstream of ALK exon 20 ( FIGS. 1B and 6A -C).
  • the novel ALK transcript has a size of approximately 2500 bps ( FIG. 7A ).
  • the ALK ATI transcript has three predicted in-frame start codons (ATGs) resulting in proteins with molecular weights (MW) of 61.1 kDa (552 amino acids), 60.8 kDa (550 amino acids) and 58.7 kDa (532 amino acids) ( FIGS. 6D and 6E ). All three proteins maintain the intracellular tyrosine kinase domain, but lack the extracellular and transmembrane domains of ALK WT ( FIG. 2A ).
  • Immunoblots of two ALK WT -expressing neuroblastoma cell lines and two EML4-ALK variant-expressing lung cancer cell lines showed proteins at the expected MW of ⁇ 220 kDa for ALK WT (and a smaller cleavage product lacking part of the extracellular region 49 ) and of ⁇ 120 kDa and ⁇ 90 kDa for two EML4-ALK variants.
  • ALKATI-expressing tumors revealed a double band at ⁇ 60 kDa, suggesting that ALKATI is translated from more than one start codon ( FIG. 2B ).
  • the three start codons were mutated individually or in combination and expressed them in 293T cells. Immunoblots revealed that each of the mutant ALK ATI lost the corresponding protein band, indicating that all three start codons in ALK ATI are functional and give rise to three distinct proteins ( FIG. 2C ).
  • ALK ATI proteins were phosphorylated in tumors with endogenous ALK ATI expression and in cells with exogenous ALK ATI expression ( FIGS. 2B and 2C ) indicating that ALK ATI is active. This was confirmed by an in vitro kinase assay ( FIG. 12A ).
  • ALK ATI may auto-activate by forming homodimers as do other RTKs 50 .
  • the ability of self-interaction was tested using co-immunoprecipitation with V5- or HAtagged ALK ATI proteins.
  • the V5-ALK ATI readily co-immunoprecipitated with the HA-ALK ATI and vice versa, indicating that ALK ATI self-interacts resulting in autophosphorylation and kinase activation ( FIG. 2D ).
  • ALK ATI was detected in both the nucleus and the cytoplasm, whereas AL KF1174L and EML4-ALK were found in the cytoplasm or the cell membrane ( FIG. 2E ).
  • ALK immunohistochemistry in clinical samples confirmed the nuclear and cytoplasmic localization of ALK ATI , suggesting that nuclear ALK staining in immunohistochemistry could serve as a clinical biomarker to identify patients with ALK ATI -expressing tumors ( FIGS. 2F and 12B ).
  • ALK ATI is not expressed in normal tissues.
  • Ba/F3, NIH-3T3, and melan-a cells were stably transduced with ALK ATI , negative controls (ALK ATI , empty vector), and positive controls (the oncogenic ALK variants ALK F1174L , EML4-ALK, and ALK WT , which was previously shown to be sufficient to drive oncogenesis at high endogenous expression levels 6,12,58-61 ).
  • ALK ATI expression led to IL-3-independent cell growth, as did the positive controls, but not the negative controls ( FIG. 3A ).
  • ALK-transformed Ba/F3 cells growing in media without IL-3 it was confirmed that ALK ATI was expressed at a similar level compared to human tumors and that all ALK isoforms were phosphorylated and therefore active ( FIG. 3B ).
  • the ALK dependency of IL-3-independent growth was reflected in the selection of Ba/F3 cells expressing greenfluorescent-protein (GFP), which was co-expressed from the ALK expression vectors ( FIG. 12C ).
  • GFP greenfluorescent-protein
  • ALK-variant expressing cells (ALK ATI , ALK F1174L , EML4-ALK, ALK WT ) were able to establish growth factor-independent proliferation and tumorigenesis, with similar growth rates once the tumors were established.
  • ALK WT the observed oncogenic capacity of ALK WT is consistent with previous reports that high endogenous expression or amplification of ALK WT drives oncogenesis and confers sensitivity to ALK inhibitors in neuroblastomas 10,12, 58-61 .
  • NIH-3T3 cells were stably transduced with either a low or high titre of ALK ATI resulting in cells expressing ALK ATI at either low or at high levels. It was found that a further increase in ALK ATI expression levels did not accelerate tumor graft formation and tumor growth indicating that ALKATI can drive tumourigenesis once a threshold of expression is reached ( FIGS. 12G-12I ).
  • Ba/F3 cells stably expressing various ALK isoforms were treated with three different ALK inhibitors (crizotinib, ceritinib, TAE-684). All ALK inhibitors effectively inhibited IL-3-independent growth of ALK-transformed Ba/F3 cells, whereas they had no effect on growth in the presence of IL-3 ( FIGS. 4A and 13A-13B ). Crizotinib inhibited ALK ATI phosphorylation and downstream signaling in a concentration-dependent manner, further corroborating that ALK ATI is activated through auto-phosphorylation ( FIGS. 4B and 13E-13E ).
  • FIGS. 4F and 4G a patient with metastatic melanoma with ALK ATI expression was identified.
  • the patient had previously progressed on the combination of ipilimumab and nivolumab immunotherapy in a clinical trial, followed by palliative radiation and then dacarbazine chemotherapy.
  • the compassionate use of crizotinib resulted in marked symptomatic improvement and tumor shrinkage within 6 weeks of therapy ( FIG. 4H ).
  • This patient case provides additional evidence that ALK ATI confers gain-of-function in patients and is amenable to pharmacologic targeting, warranting further clinical investigation.
  • ALK ATI a novel ALK transcript, which arises independently of genomic aberrations at the ALK locus through alternative transcriptional initiation, was identified.
  • ALK ATI encodes shortened ALK proteins that are capable of driving oncogenesis in vitro and in vivo.
  • ALK ATI -driven tumors are sensitive to ALK inhibitors, suggesting that patients harboring such tumors could potentially benefit from ALK inhibitor therapy, but may not be identified using current clinical genomic assays, particularly those based on DNA sequencing.
  • alternative transcriptional initiation was discovered as a novel mechanism for oncogene activation, in addition to well-established genetic mechanisms such as mutations, translocations or amplifications. Other oncogenes may be activated via similar mechanisms in other human malignancies and their identification may provide new insights into oncogenesis and opportunities for therapeutic intervention.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Epidemiology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Analytical Chemistry (AREA)
  • Pathology (AREA)
  • Urology & Nephrology (AREA)
  • Cell Biology (AREA)
  • Hematology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Hospice & Palliative Care (AREA)
  • Oncology (AREA)
  • Physics & Mathematics (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Biophysics (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to a Truncated isoform of Anaplastic Lymphoma Kinase (“TALK”). Expression of this isoform is associated with malignancy and with responsiveness to ALK inhibitors. Detection of the isoform may be used in diagnostic and therapeutic methods. Because it arises as a result of variant transcription rather than genetic rearrangement, its presence would be undetected by genomic testing.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of PCT International Patent Application No. PCT/US2015/025289, filed on Apr. 10, 2015, which claims the benefit of U.S. Provisional Patent Application Ser. No. 61/978,106 filed Apr. 10, 2014, the contents of each of which are hereby incorporated by reference in their entirety herein, and to each of which priority is claimed.
  • SEQUENCE LISTING
  • The specification further incorporates by reference the Sequence Listing submitted herewith via EFS on Oct. 7, 2016. Pursuant to 37 C.F.R. § 1.52(e)(5), the Sequence Listing text file, identified as 072734.0241USSEQ.txt, is 78,868 bytes and was created on Oct. 7, 2016. The Sequence Listing, electronically filed herewith, does not extend beyond the scope of the specification and thus does not contain new matter.
  • 1. INTRODUCTION
  • The present invention relates to a Truncated isoform of Anaplastic Lymphoma Kinase (“TALK”). Expression of this isoform is associated with malignancy and with responsiveness to ALK inhibitors. Detection of the isoform may be used in diagnostic and therapeutic methods. Because it arises as a result of variant transcription rather than genetic rearrangement, its presence would be undetected by genomic testing.
  • 2. BACKGROUND OF THE INVENTION
  • Comprehensive characterization of genetic aberrations underlying human cancer is essential for improving tumor diagnostics, identifying therapeutic targets, developing rational combination therapies and optimizing clinical trial designs3. Large scale sequencing studies, such as The Cancer Genome Atlas (TCGA) project, continue to reveal an increasingly detailed picture of the genetic aberrations in many cancer types, but focus largely on characterizing genetic aberrations in the coding regions of DNA2-5. However, in a significant proportion of tumors, the number of detectable genetic aberrations in driver oncogenes is too low to explain malignant transformation6. This is exemplified by a recent study, in which ˜15% of lung adenocarcinoma lacked gene mutations affecting any of the hallmarks of cancer7,8. These observations suggest that mechanisms other than genetic aberrations may be involved in malignant transformation.
  • 3. SUMMARY OF THE INVENTION
  • The present invention relates to a truncated isoform of anaplastic lymphoma kinase, also referred to as “TALK” herein, and its use in diagnosis and treatment of cancer patients. It is based, at least in part, on the discovery of a novel oncogenic TALK, termed “ALKATI”, that arose as a result of the establishment of a de novo alternative transcriptional initiation (ATI) site in ALK intron 19 independent of genetic aberrations at the ALK locus. The ALKATI transcript encodes three proteins with a molecular weight of 61.1, 60.8 and 58.7 kDa consisting primarily of the intracellular tyrosine kinase domain. ALKATI was found to stimulate multiple oncogenic signaling pathways, drive growth factor-independent cell proliferation in vitro, and promote tumourigenesis in vivo. ALK inhibitors were found to suppress the kinase activity of ALKATI, suggesting that patients with ALKATI-expressing tumors may benefit from ALK inhibitors. Expression of ALKATI was found in more than 10% (˜11%) of melanomas and sporadically in other cancer types, but not in normal tissues. Detection of TALK, e.g., ALKATI in a cell of a subject may be used to diagnose a cancer in the subject and may be used to determine whether treatment of the subject with an ALK inhibitor would be more likely to confer therapeutic benefit. It is an advantage of the invention that a subject expressing TALK who would appear normal by genomic testing may be identified by detecting TALK transcription, mRNA or protein.
  • The present invention provides an isolated nucleic acid molecule comprising exons 20-29 and a portion but not all of intron 19 of an Anaplastic Lymphoma Kinase (ALK) gene, where the nucleic acid molecule does not comprise exons 1-19 of the ALK gene, either individually or in any combination. In certain embodiments, the ALK gene is a human ALK gene. In one non-limiting embodiment, the portion of intron 19 of the ALK gene is about 400 bp in length located upstream of exon 20 of the ALK gene. In certain embodiments, the nucleic acid molecule is about 2300-2600 bp in length, such as about 2500 bp in length, e.g., about 2513 bp in length. In one non-limiting embodiment, the nucleic acid molecule is a messenger RNA (mRNA) transcript. In certain embodiments, the isolated nucleic acid molecule comprises the nucleotide sequence set forth in SEQ ID NO: 11 or a sequence that is at least about 95% homologous thereto.
  • The present invention also provides an isolated nucleic acid molecule consisting essentially of exons 20-29 and a portion but not all of intron 19 of an Anaplastic Lymphoma Kinase (ALK) gene, wherein the nucleic acid molecule does not comprise exons 1-19 of the ALK gene, either individually or in any combination. In certain embodiments, the isolated nucleic acid molecule consists essentially of the nucleotide sequence set forth in SEQ ID NO: 11 or a sequence that is at least about 95% homologous thereto.
  • The present invention further provides an isolated complementary DNA (cDNA) molecule comprising exons 20-29 of an Anaplastic Lymphoma Kinase (ALK) gene, wherein the cDNA molecule does not comprise exons 1-19 of the ALK gene, either individually or in any combination. In certain embodiments, the ALK gene is a human ALK gene. In certain embodiments, the isolated cDNA molecule comprises nucleic acids 405-2063 of the nucleotide sequence set forth in SEQ ID NO: 11 or a sequence that is at least about 95% homologous thereto. In certain embodiments, the isolated cDNA molecule comprises nucleic acids 411-2063 of the nucleotide sequence set forth in SEQ ID NO: 11 or a sequence that is at least about 95% homologous thereto. In certain embodiments, the isolated cDNA molecule comprises nucleic acids 465-2063 of the nucleotide sequence set forth in SEQ ID NO: 11 or a sequence that is at least about 95% homologous thereto.
  • Additionally, the present invention provides an isolated complementary DNA (cDNA) molecule consisting essentially of exons 20-29 of an Anaplastic Lymphoma Kinase (ALK) gene, wherein the cDNA molecule does not comprise exons 1-19 of the ALK gene, either individually or in any combination. Also provided is an isolated cDNA molecule consisting essentially of nucleic acids 405-2063 of the nucleotide sequence set forth in SEQ ID NO: 11 or a sequence that is at least about 95% homologous thereto, wherein the cDNA molecule does not comprise exons 1-19 of the ALK gene, either individually or in any combination. The present invention further provides an isolated cDNA molecule consisting essentially of nucleic acids 411-2063 of the nucleotide sequence set forth in SEQ ID NO: 11 or a sequence that is at least about 95% homologous thereto, wherein the cDNA molecule does not comprise exons 1-19 of the ALK gene, either individually or in any combination. The present invention further provides an isolated cDNA molecule consisting essentially of nucleic acids 465-2063 of the nucleotide sequence set forth in SEQ ID NO: 11 or a sequence that is at least about 95% homologous thereto, wherein the cDNA molecule does not comprise exons 1-19 of the ALK gene, either individually or in any combination.
  • The present invention provides an isolated nucleic acid molecule comprising a nucleotide sequence encoding the amino acid sequence set forth in SEQ ID NO: 12 or a sequence that is at least about 95% homologous thereto; an isolated nucleic acid molecule comprising a nucleotide sequence encoding amino acids 3 to 552 of the amino acid sequence set forth in SEQ ID NO: 12 or a sequence that is at least about 95% homologous thereto; or an isolated nucleic acid molecule comprising a nucleotide sequence encoding amino acids 21 to 552 of the amino acid sequence set forth in SEQ ID NO: 12 or a sequence that is at least about 95% homologous thereto.
  • The present invention also provides a vector comprising the above-described isolated nucleic acid molecule, or the above-described isolated cDNA molecule. The invention provides a host cell comprising such vector. Furthermore, the invention provides an isolated polypeptide encoded by the above-descried nucleic acid molecule, or an isolated polypeptide encoded by the above-described cDNA molecule.
  • The present invention further provides an isolated polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 12 or a sequence that is at least about 95% homologous thereto, where the polypeptide does not comprise the amino acid sequence set forth in SEQ ID NO; 3. Also provided is an isolated polypeptide comprising amino acids 3 to 552 of the amino acid sequence set forth in SEQ ID NO: 12 or a sequence that is at least about 95% homologous thereto, where the polypeptide does not comprise the amino acid sequence set forth in SEQ ID NO; 3. The present invention further provides an isolated polypeptide comprising amino acids 21 to 552 of the amino acid sequence set forth in SEQ ID NO: 12 or a sequence that is at least about 95% homologous thereto, where the polypeptide does not comprise the amino acid sequence set forth in SEQ ID NO; 3.
  • Additionally, the present invention provides an isolated polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 12 or a sequence that is at least about 95% homologous thereto, where the polypeptide does not comprise the amino acid sequence set forth in SEQ ID NO; 4. Also provided is an isolated polypeptide comprising amino acids 3 to 552 of the amino acid sequence set forth in SEQ ID NO: 12 or a sequence that is at least about 95% homologous thereto, where the polypeptide does not comprise the amino acid sequence set forth in SEQ ID NO; 4. The present invention further provides an isolated polypeptide comprising amino acids 21 to 552 of the amino acid sequence set forth in SEQ ID NO: 12 or a sequence that is at least about 95% homologous thereto, where the polypeptide does not comprise the amino acid sequence set forth in SEQ ID NO; 4.
  • The invention also provides an antibody that binds to any of the above-described polypeptides, wherein the antibody does not bind to a wild type ALK. In certain embodiments, the wild type ALK is a wild type human ALK, e.g., one comprising the amino acid sequence set forth in SEQ ID NO: 1.
  • The present invention also provides a method of diagnosing a cell as a cancer cell comprising detecting the presence of a Truncated isoform of Anaplastic Lymphoma Kinase (TALK) in the cell, where the presence of the TALK indicates that the cell is a cancer cell. In certain embodiments, the method comprises determining the presence of a detectable TALK mRNA transcript, a detectable TALK cDNA molecule corresponding thereto, a detectable TALK polypeptide encoded thereby, and/or a detectable alternative transcriptional initiation (ATI) site in intron 19 of an ALK gene in the cell.
  • The present invention provides a method of determining whether an anti-cancer effect is likely to be produced in a cancer by an ALK inhibitor, comprising determining whether one or more cell of the cancer contains a detectable TALK, wherein the presence of a detectable TALK in the cell indicates that an ALK inhibitor would have an anti-cancer effect on the cancer. In certain embodiments, the method comprises determining the presence of a detectable TALK mRNA transcript, a detectable TALK cDNA molecule corresponding thereto, a detectable TALK polypeptide encoded thereby, and/or a detectable ATI site in intron 19 of an ALK gene in the cell.
  • The present invention further provides a method of treating a subject having a cancer comprising: (a) determining whether a subject is likely to obtain therapeutic benefit from an ALK inhibitor, comprising determining whether one or more cancer cell of the subject contains a detectable TALK, wherein the presence of a detectable TALK in the cancer cell of the subject indicates that the subject is likely to benefit from an ALK inhibitor; and (b) treating the subject who is likely to benefit from an ALK inhibitor with a therapeutic amount of an ALK inhibitor. In certain embodiments, the method comprises treating the subject with an alternative therapy other than an ALK inhibitor if no detectable TALK is present in the cancer cell of the subject. In certain embodiments, the method further comprises obtaining a sample of one or more cancer cell of the subject before treating the subject with an ALK inhibitor. In certain embodiments, the subject who is likely to benefit from an ALK inhibitor receives an ALK inhibitor and one or more additional cancer treatment selected from the group consisting of one or more BRAF inhibitor, one or more MEK inhibitor, one or more immunologic inhibitor, one or more CDK4 inhibitor, one or more CDK6 inhibitor, one or more alklyating agent, one or more topoisomerase inhibitor, one or more anti-metabolite, one or more anti-microtubule agent, one or more cytotoxic antibiotic, radiation therapy, chemotherapy, and combinations thereof. In certain embodiments, the immunologic inhibitor is selected from the group consisting an anti-PD-1 antibody, an anti-CTLA-4 antibody, an anti-PD-L1 antibody. In one non-limiting embodiment, the subject who is likely to benefit from an ALK inhibitor receives an ALK inhibitor, an anti-CTLA-4 antibody, and an anti-PD-1 antibody. In one non-limiting embodiment, the subject further receives radiation and chemotherapy. In certain embodiments, the method comprises determining the presence of a detectable TALK mRNA transcript, a detectable TALK cDNA molecule, a detectable TALK polypeptide encoded thereby, and/or a detectable ATI site in intron 19 of an ALK gene in the cell.
  • The present invention also provides a kit for determining whether an anti-cancer effect is likely to be produced in a cancer by an ALK inhibitor, comprising a means for determining the presence of a detectable TALK in one or more cell of the cancer. In certain embodiments, the kit comprises means for determining the level of a TALK mRNA transcript, a TALK cDNA molecule corresponding thereto, and/or a TALK polypeptide encoded thereby. In certain embodiments, the means for determining the level of a TALK mRNA transcript is selected from the group consisting of probe hybridization, polymerase chain reaction (PCR), Northern blot, sequencing, microarray, and combinations thereof. In certain embodiments, the kit comprises a nucleic acid probe that hybridizes with the TALK mRNA transcript to determine the level of the TALK mRNA transcript. In certain embodiments, the probe hybridization is a probe-based NanoString nCounter assay. In certain embodiments, the kit comprises a color coded probe pair that hybridizes with the TALK mRNA transcript to determine the level of the TALK mRNA transcript. In certain embodiments, the PCR is selected from the group consisting of reverse transcription polymerase chain reaction (RT-PCR), quantitative reverse transcriptase PCR, real-time PCR, quantitative real-time PCR, and combinations thereof. In certain embodiments, the kit comprises a pair of nucleic acid primers that hybridizes with the TALK mRNA to determine the level of the TALK mRNA transcript. In certain embodiments, the means for determining the level of a TALK cDNA molecule is PCR. In certain embodiments, the kit comprises a pair of nucleic acid primers that hybridize with the TALK cDNA molecule to determine the level of the TALK cDNA molecule. In certain embodiments, the means for determining the level of a TALK polypeptide is selected from the group consisting of antibody binding, immunohistochemistry, Western blot, a functional assay, enzyme linked immunosorbent assay (ELISA), radioimmunoassays (RIA), enzyme immunoassays (EIA), mass spectrometry, a 1-D or 2-D gel-based analysis system, immunoprecipitation, and combinations thereof. In certain embodiments, the kit comprises an antibody or a fragment thereof that specifically binds to the TALK polypeptide. In one non-limiting embodiment, the functional assays is a kinase assay.
  • In certain embodiments, the kit comprises means for determining the presence of an ATI site in intron 19 of an ALK gene in one or more cell of the cancer. In certain embodiments, the means for determining the presence of an ATI site in intron 19 of an ALK gene is selected from the group consisting of Northern blot, Chromatin immunoprecipitation (ChIP)-seq, ChIP-qPCR, Rapid Amplification of cDNA Ends (RACE)-PCR, and combinations thereof.
  • The present invention provides a means for determining the presence of a detectable TALK for use in a method of determining whether an anti-cancer effect is likely to be produced in a cancer by an ALK inhibitor, the method characterized by determining whether one or more cell of the cancer contains a detectable TALK, where the presence of a detectable TALK in the cell indicates that an ALK inhibitor would have an anti-cancer effect on the cancer. In certain embodiments, the means is for determining the level of a TALK mRNA transcript in one or more cell of the cancer. In certain embodiments, the means is selected from the group consisting of probe hybridization, polymerase chain reaction (PCR), Northern blot, sequencing, microarray, and combinations thereof. In one non-limiting embodiment, the probe hybridization is a probe-based NanoString nCounter assay. In certain embodiments, the PCR is selected from the group consisting of reverse transcription polymerase chain reaction (RT-PCR), quantitative reverse transcriptase PCR, real-time PCR, quantitative real-time PCR, and combinations thereof. In certain embodiments, the means is for determining the level of a TALK cDNA molecule in one or more cell of the cancer. In one non-limiting embodiment, the means is PCR. In certain embodiments, the means is for determining the level of a TALK polypeptide in one or more cell of the cancer. In certain embodiments, the means is selected from the group consisting of antibody binding, immunohistochemistry, Western blot, a functional assay, enzyme linked immunosorbent assay (ELISA), radioimmunoassays (RIA), enzyme immunoassays (EIA), mass spectrometry, a 1-D or 2-D gel-based analysis system, immunoprecipitation, and combinations thereof. In one non-limiting embodiment, the functional assays is a kinase assay. In certain embodiments, the means is for determining the presence of an ATI site in intron 19 of an ALK gene in one or more cell of the cancer. In certain embodiments, the means is selected from the group consisting of Northern blot, Chromatin immunoprecipitation (ChIP)-seq, ChIP-qPCR, Rapid Amplification of cDNA Ends (RACE)-PCR, and combinations thereof.
  • The present invention further provides an ALK inhibitor for use in treating a subject having cancer, wherein the ALK inhibitor is more likely to produce an anti-cancer effect if one or more cancer cell of the subject contains a detectable TALK.
  • In certain embodiments, the cancer is selected from the group consisting of melanoma, thyroid carcinoma, lung adenocarcinoma, lung squamous cell carcinoma, renal clear cell carcinoma, and breast cancer. In one non-limiting embodiment, the cancer is melanoma. In one non-limiting embodiment, the cancer is anaplastic thyroid carcinoma.
  • In certain embodiments, the ALK inhibitor is selected from the group consisting of crizotinib, ceritinib, NVP-TAE684, alectinib; AP26113, ASP-3026, CEP-37440, NMS-E628, PF-06463922, TSR-011, RXDX-101, and X-396. In one non-limiting embodiment, the ALK inhibitor is crizotinib.
  • In certain embodiments, the TALK mRNA transcript is the above-described nucleic acid molecule. In certain embodiments, the TALK cDNA molecule is the above-described cDNA molecule. In certain embodiments, the TALK polypeptide is the above-described polypeptide.
  • 4. BRIEF DESCRIPTION OF THE FIGURES
  • FIGS. 1A-1H. Alternative transcriptional initiation (ATI) results in a novel ALK transcript. 1A, Distribution of RNA-seq reads of ALK variant transcripts: ALKATI RNAseq reads aligned to both ALK intron 19 and exons 20-29; full-length ALKWT RNA-seq reads aligned to all ALK exons, but not to the introns; translocated ALK RNA-seq reads aligned only to ALK exons 20-29. 1B, Mapping of the ATI sites of ALKATI by 5′-RACE and next-generation sequencing. More than 95% of the ALKATI transcripts start within a 25 base-pair region in ALK intron 19 (hg19 ch2:29,446,768-29,446,744; blue shaded area). 1C, ChIP-seq profile of H3K4me3 at the ATI site of an ALKATI-expressing tumor (MM-15), a melanoma cell line (SKMEL-524) without ALKATI expression, and a lung cancer cell line (H3122) with a EML4-ALK fusion. The shape of the peak shows a slow taper towards the gene body characteristic of active promoters. 1D, ChIP-qPCR validation of H3K4me3 binding at the ATI site. Mean±SEM, n=3. 1E, ChIP-qPCR of RNA-pol II binding at the ATI site. Mean±SEM, n=3. 1F, Quantitative mRNA profiling of different ALK variants using Nanostring nCounter. Two ALKWT-expressing neuroblastoma cell lines (SK-N-BE2, SK-N-DZ), two lung cancer cell lines (H3122, H2228) with EML4-ALK translocations, nine ALKATI expressing tumors: 8 melanomas (MM) and 1 anaplastic thyroid carcinoma (ATC). 1G, Similar SNV frequencies in DNA-seq, RNA-seq and ChIP-seq (H3K4me3) data indicate that ALKATI is bi-allelically expressed and that both ALK alleles are decorated with H3K4me3. 1H, Table showing the probe set used for the Nanostring nCounter Assay to quantify the mRNA levels of ALKATI, ALKWT, and translocated ALK.
  • FIGS. 2A-2F. The ALKATI transcript encodes three shortened ALK proteins containing mainly the ALK kinase domain. 2A, Illustration of ALK protein isoforms. ALK, wildtype (ALKWT) consists of a signaling peptide from amino acid (aa) position 1-18, an extracellular segment with two MAM domains (meprin, A-5 protein, and receptor protein-tyrosine phosphatase mu) and one glycine rich region (Gly), a transmembrane segment, a juxtamembrane segment, and an intracellular segment with the tyrosine kinase domain (kinase, aa 1116-1392). Activating ALK mutations occur usually in the kinase domain, such as ALKF1174L. The EML4-ALK fusion protein and ALKATI contain the entire kinase domain and parts of the juxtamembrane segment. The translation of ALKATI initiates at one of three in-frame start codons (ATGs) as indicated. 2B, Immunoblots of total- (t-) and phosphorylated- (p-) ALK in two ALKWT-expressing neuroblastoma cell lines (SK-N-DZ, SK-N-BE2), two lung cancer cell lines (H3122, H2228) expressing different variants of EML4-ALK fusion, three tumor samples expressing ALKATI (MM-15, MM-74, ATC-28) and a negative control melanoma cell line (SKMEL-28). 2C, Immunoblots of 293T cells transduced with ALKATI, in which the three predicted start codons were mutated from ATG to AAG, individually or in combination as indicated. 2D, Co-immunoprecipitation (IP) and immunoblots (IB) of the indicated epitopes in 293T cells with exogenous expression of V5-tagged ALKATI (V5-ALKATI) or HA-tagged ALKATI (HA-ALKATI), or both, demonstrating that ALKATI proteins self-interact. 2E, ALK immunofluorescence in NIH-3T3 cells expressing the indicated ALK isoforms. Scale bar, 25 μm. 2F, HE (haematoxylin-eosin) stain and ALK immunohistochemistry in ALKATI-expressing human tumor samples. Scale bar, 50 μm.
  • FIGS. 3A-3C. ALKATI promotes growth factor-independent proliferation in vitro and tumourigenesis in vivo. 3A, Growth curves of Ba/F3 cells stably expressing the indicated ALK isoforms in the absence of interleukin 3 (IL-3). Mean±SEM, n=4. 3B, Immunoblots to determine the ALK levels in Ba/F3 cells. Cells were previously transformed by the expression of different ALK isoforms and selected for growth in the absence of IL-3. All ALK variants were phosphorylated when expressed at levels required for IL-3-independent growth. 3C, Growth curves of tumor grafts of NIH-3T3 cells stably expressing the indicated ALK isoforms. ALKF1174L, EML4-ALK, and overexpression and amplification of ALKWT are well-established oncogenic drivers in various tumors. Mean±SEM, n=8.
  • FIGS. 4A-4H. ALKATI-expression confers sensitivity to ALK inhibitors in vitro and in vivo. 4A, Dose-response curves for the ALK inhibitor crizotinib in Ba/F3 cells expressing the indicated ALK isoforms in the presence or absence of IL-3. Mean±SEM, n=3. 4B, Representative immunoblots of Ba/F3 cells expressing ALKATI and treated with increasing concentrations of crizotinib for 2 hours. 4C, Normalised tumor volume over time in SCID mice implanted with NIH-3T3 cells expressing the indicated ALK isoforms and treated with either vehicle or crizotinib (100 mg/kg/day). Mean±SEM, n=8. 4D, Haematoxylineosin (HE) staining and immunohistochemistry (IHC) of explanted ALKATI-expressing tumors 48 hours after first crizotinib treatment. Scale bar, 50 μm. 4E, Normalised bioluminescence of luciferase-labelled NIH-3T3 grafted tumors expressing ALKATI over time in SCID mice treated with either vehicle or crizotinib (100 mg/kg/day). Mean±SEM, n=4. 4F, HE staining and ALK-IHC (insert) of the melanoma metastasis from patient 1 (MM-382). 4G, Quantitative mRNA profiling of ALKATI using Nanostring nCounter. Controls expressing ALKWT, EML4-ALK or ALKATI compared to the melanoma metastasis of patient MM-382. Scale bar, 50 μm. 4H, Computed tomography (CT) images of a representative subcutaneous melanoma metastasis on the left hip of patient 1 (MM-382) before and after crizotinib treatment, respectively.
  • FIGS. 5A-5C. Comparison of the RNA seq profiles of various ALK transcript. RNA-seq data are displayed in the Integrative Genomics Viewer (IGV). The grey bars/arrows indicate the sequencing reads. The blue lines connect sequencing reads that are aligned over the splice site of joining exons. 5A, The ALKATI transcript shows expression of ALK exons 20-29 and of approx. 400 bps in intron 19 (blue shaded area). No expression of exon 1-19 or intronic areas, other than in intron 19, is observed. The detailed view illustrates that the sequencing reads align continuously between exon 20 and intron 19 indicating uninterrupted transcription. The 5′-UTR of ALKATI (intron 19) and exon 20-29 are expressed at comparable levels. 5B, The full-length ALKWT transcript shows expression of all ALK exons and only very little expression of the introns. The detailed view displays that the sequencing reads align sharply to the exons, but not to the intron 19 region, which is present in ALKATI (blue shaded area). 5C, The ALK fusion transcript of a non-small cell lung cancer with an EML4-ALK translocation shows expression of ALK exons 20-29, and little expression of exons 1-19 and all introns. The detailed view illustrates that the transcription starts mainly at exon 20 due to the preserved splice site. Only few reads are aligned to the intron 19 region (blue shaded area). The green-labelled reads highlight chimeric read pairs indicating the EML4-ALK translocation.
  • FIGS. 6A-6G. Identification of the novel ALKATI transcript. 6A, IGV view of the 5′-RACE-cDNA fragments obtained by massively parallel sequencing. The vast majority of sequencing reads (grey arrows) start within the main ATI site of 25 base pairs (hg19 chr2:29,446,744-768). 6B, Percentage of reads starting at the ATI site in ALKATI-expressing tumor samples. 6C, Sanger sequencing of the cloned 5′-RACE-cDNA fragments confirm the continuous transcription starting in ALK intron 19 and extending to exons 20-21. 6D, The ALKATI transcript consists of approximately 400 bp upstream of exon 20 and of ALK exons 20-29. The transcriptional initiation site was defined as the first base pair at which more than 5% of the transcripts were initiated (chr2:29,446,766). Other major transcriptional initiation sites are marked in red, the 5′- and 3′-UTRs in dark blue, the coding DNA sequence (CDS) in black and in box, and the first and last base of each exon in light blue. The translation is initiated at 3 start codons (ATGs; bold and underlined): 1st ATG: hg19 chr2:29,446,360-2, 2nd ATG (+7-9), and 3rd ATG (+61-3). 6E, The amino acid sequence of ALKATI. The translation is initiated at one of 3 start codons. The corresponding 3 methionines (bold and underlined) result in 3 different proteins, 61.08 kDa (552 amino acids), 60.82 kDa (550 amino acids) and 58.71 kDa (532 amino acids). The kinase domain is highlighted in red letters. The lysine in the ATP binding domain in ALKWT is marked bold and underlined, and was mutated to methionine (p.K1150M) in the kinase-dead ALKATI-KD. 6F, Northern blot of full-length ALKWT (neuroblastoma cell lines), EML4-ALK (lung cancer cell lines; variant 1 and 3), ALKATI from human melanomas, and negative controls (melanoma cell lines). Except for the negative controls, each lane shows two bands; the lower V2 band matches the shorter canonical (RefSeq) ALK transcript ending at ˜chr2:29,415,640; the upper V1 band corresponds to a transcript with a 1.8 kb longer 3′ UTR ending at ˜chr2:29,413,840. Two ALKATI expressing tumors, MM-284 and MM-74, show only weak signals because less than 1 μg RNA were available; for all other samples 5-10 μg RNA were used. 6G, RNA-seq Sashimi plot illustrating the shorter V2 and the longer V1 ALK transcripts by the sharp drop of sequencing reads in the 3′ UTR at chr2:29,415,640 for V2 and at chr2:29,413,840 for V1.
  • FIGS. 7A-7D. RNApol II and H3K4me3 are enriched at the ATI site of ALKATI-expressing tumor samples. 7A, Primer sequences used for ChIP-qPCR. 7B, Schematics of the ChIP-qPCR primer binding sites at the ALKATI locus. 7C and 7D, ChIP-qPCR of (C) H3K4me3 and (D) RNApol II at the ATI site demonstrating enrichment of both marks in the ALKATI-expressing human tumor samples, but not in the negative controls, including a lung cancer cell line with EML4-ALK translocation (H3122), and a melanoma cell line (SKMEL-524). Results for primer pair 1 (P1) are shown in FIGS. 1D-1E. Mean±SEM, n=3.
  • FIGS. 8A-8E. ALKATI-expressing tumors in the TCGA dataset. 8A, The frequency of ALKATI-expressing tumors in more than 5,000 tumor samples from 15 different cancer types in the TCGA RNA-seq dataset. 8B-8E, IGV views of the ALK locus of representative ALKATI-expressing tumor types in the TCGA dataset including B, melanoma; C, lung adenocarcinoma; D, breast invasive carcinoma; E, clear cell renal cell carcinoma. The absence of chimeric sequencing read pairs indicates no translocations in these cases.
  • FIGS. 9A-9D ALKATI is transcribed from a genomically intact ALK locus. 9A, Interphase FISH with ALK flanking probes demonstrates juxtaposed green and orange signals indicating no ALK rearrangement in MM-15. Scale bar, 10 μm. 9B, Interphase FISH shows no ALK rearrangement, but 3 green/orange fusion signals in the majority of cell nuclei indicating a trisomy 2 in MM-74. Scale bar, 10 μm. 9C, the top panel shows the genome-wide array CGH profile of MM-15 with numerous chromosomal gains and losses across the entire genome, which are characteristic of metastatic melanoma. The chromosomes are aligned along the x-axis. The blue line illustrates the relative copy number (log 2 ratio) and the blue bars highlight copy number gains and losses. The middle panel illustrates the relative copy number (blue line) across chromosome 2. Distal to the ALK locus, a loss on the p-arm of chromosome 2 is indicated. The lower panel illustrates the relative copy number across the ALK gene. The red and green squares represent the log 2 ratio of individual aCGH probes (green: positive log 2 ratio; red: negative log 2 ratio). No disruption or selective gains or losses are found at the ALK locus. 9D, The genome-wide array CGH profile of MM-74 shows numerous chromosomal gains and losses across the entire genome in the top panel. The middle panel displays a relative copy number gain of the entire chromosome 2, which is in line with the trisomy of chromosome 2 as indicated by FISH. The lower panel also displays trisomy of chromosome 2 and shows no focal gains and losses at the ALK locus.
  • FIGS. 10A-10D. Targeted sequencing and whole-genome sequencing reveals no recurrent genomic aberrations at the ALK locus. 10A, Data from ultra-deep sequencing of the entire ALK locus are displayed in IGV. The genomic region around intron 19 reveals several single nucleotide variations (SNVs). However, the vast majority of SNVs at the ALK locus are also found in the general population as they are detected in the pool of normal DNA, which was used as the control (pooled normal, bottom panel). Numerous SNVs are also documented in The Single Nucleotide Polymorphism database (dbSNP—www.ncbi.nlm.nih.gov/SNP/). No genomic aberrations were found at the transcriptional initiation site of ALKAT. 10B and 10C, Circos plots of the whole-genome sequencing data of (B) MM-15 and (C) ATC-28 illustrating numerous SNV and structural aberrations. 10D, Single nucleotide polymorphisms and small indels at the ALK locus detected by ultra-deep sequencing.
  • FIGS. 11A-11H. Local chromatin context at the alternative transcription initiation (ATI) site. 11A, UCSC Genome Browser view at the ATI site. The RepeatMasker track shows transposable elements at the ATI region, including a long-terminal repeat (LTR) in intron 19 (LTR16B2) and a long-interspaced element (LINE) in intron 18. The ENCODE tracks reveal a DNase I hypersensitivity cluster and H3K4me1 enrichment, but no H3K27ac enrichment. 11B, The methylation status of the ALK locus was assessed by custom capture of the entire ALK footprint, followed by bisulfite treatment and next-generation sequencing. Bisulfite sequencing results of H3122 and MM-15 are displayed in the CG-bisulfite mode of IGV. The red color denotes “C” (Cytosine) corresponding to methylated cytosine, which is preserved during the bisulfite reaction. The blue color denotes “T” (thymine) corresponding to un-methylated cytosine, which is converted to uracil in the bisulfite reaction, and subsequently amplified to thymine during PCR. 11C, Methylation level at CpGs in ALKATI-expressing tumor samples (MM-15 and ATC-28) and non-ALKATI-expressing control cells (H3122, a lung cancer cell line with EML4-ALK expression and SKMEL-28, a melanoma cell line without ALKATI expression) at the indicated genomic loci. Black lines mark the LTR16B2 (LTR), red lines mark the LINE. 11D, Comparison of the methylation status of CpGs adjacent to the ATI site in ALKATI-expressing tumor samples (MM-15 and ATC-28) and non-ALKATI-expressing control cells (H3122 and SKMEL-28). The regions flanking LTR16B2 have significantly lower CpG methylation levels in ALKATI-expressing samples than controls; red dots indicate a statistically significant difference (p<0.05) between ALKATI-expressing and non-expressing samples. Black dots indicate no statistically significant difference. 11E, ChIP-seq profile of H3K27ac at the ALKATI locus. The 17 blue profiles were retrieved from ENCODE, the 5 red profiles are original data from the inventors' lab. Only the three melanoma samples (MM-15, SKMEL-28, SKMEL-524; bottom), but not the 19 non-melanoma cell lines, show H3K27ac enrichment at the ATI site. 11F, ChIP-qPCR validation for the H3K27ac enrichment at the ATI site in 6 melanoma cell lines. Mean±SEM, n=3. 11G, Luciferase reporter assay of LTR16B2 in melanoma cell lines (red) and lung cancer cell lines expressing EML4-ALK (green). In contrast to the lung cancer cell lines with no luciferase activity, melanoma cell lines show low to moderate luciferase activity. 11H, Transcription factor motif analysis of the proximal cis-regulatory region (hg19 chr2: 29,445,000 to 29,447,100).
  • FIGS. 12A-12I. ALKATI is active in vitro, shows nuclear and cytoplasmic localization by immunohistochemistry, and induces tumourigenesis. 12A, In vitro kinase assay. The indicated ALK variants were stably expressed in NIH-3T3 cells, immune-precipitated, and assayed for tyrosine kinase activity. After the enzymatic reaction, the immune-precipitated material was used for immunoblots to assess the amount of ALK protein used in the kinase assay. Mean±SEM, n=4. 12B, Human tumor (MM-15) expressing ALKATI shows cytoplasmic and nuclear localization of ALK by immunohistochemistry. Melanocytic tumor expressing a TPM3-ALK translocation showed cytoplasmic localization of the ALK fusion protein without any nuclear staining by immunohistochemistry. Fibroblasts, epithelial cells and reactive lymphocytes serve as internal negative controls. Scale bars, 100 μm. 12C, Flow cytometry analysis for green-fluorescent-protein (GFP) co-expressed with the indicated ALK isoforms. Cells were cultured in IL-3 supplemented medium until day 0 (blue curve) and the number of GFP-positive was assessed. 14 days after IL-3 withdrawal, the number of GFP-positive, ALK-expressing cells was assessed again (red curve). 12D, Immunoblots of explanted NIH-3T3 tumor grafts expressing various ALK isoforms. ALKATI was expressed at similar protein levels as in two ALKATI-expressing human tumor samples. 12E, Growth curves of tumor grafts of melan-a cells stably expressing the indicated ALK isoforms. Mean±SEM, n=8. 12F, Immunoblots of explanted melan-a tumor grafts expressing various ALK isoforms in comparison to ALKATI-expressing human tumor samples. 12G, Flow cytometry analysis of the GFP signal in NIH-3T3 cells stably expressing low (ALKATI-low) or high levels of ALKATI (ALKATI-high) prior to grafting into SCID mice. 12H, Immunoblot of t-ALK in ALKATI-low and ALKATI-high cells, confirming differential expression of ATI. 12I Growth curves of tumor grafts of ALKATI-low and ALKATI-high cells.
  • FIGS. 13A-13E. Concentration dependent ALK inhibition in ALKATI, ALKWT-, ALKF1174L-, and EML4-ALK-expressing Ba/F3 cells. 13A and 13B, Cell viability assay of Ba/F3 cells, either in the presence or absence of IL-3 (1 ng/ml), expressing the indicated ALK isoforms and treated with the indicated doses of ALK inhibitors (A) ceritinib and (B) TAE-684. Cell viability was measured after 72 hours of drug treatment. Mean±SEM, n=4. 13C-13E, Representative immunoblots of Ba/F3 cells stably expressing (C) ALKWT, (D) ALKF1174L, or (E) EML4-ALK and treated with increasing concentrations of crizotinib for 2 hours. Immunoblots for ALKATI are shown in FIG. 4B.
  • FIGS. 14A-14F. Expression of ALKWT, ALKF1174L, and EML4-ALK confers sensitivity to the ALK inhibitor, crizotinib, in vivo. 14A-14C, Bioluminescence of luciferase-labelled NIH-3T3 grafted tumors expressing (A) ALKWT, (B) ALKF1174L, or (C) EML4-ALK over time in SCID mice treated with either vehicle or crizotinib (100 mg/kg/day). Data for ALKATI-expressing tumors are shown in FIG. 4E. 14D-14F, Haematoxylin-eosin staining (HE) and 25 immunohistochemistry of explanted tumors expressing (D) ALKWT, (E) ALKF1174L, or (F) EML4-ALK 48 hours after first crizotinib treatment. Data from ALKATI-expressing tumors are shown in FIG. 4D. Scale bar, 50 m.
  • 5. DETAILED DESCRIPTION OF THE INVENTION
  • For clarity and not by way of limitation, the detailed description of the invention is divided into the following subsections:
  • (i) Definitions
  • (ii) TALKs;
  • (iii) Detection of TALKs;
  • (iv) Diagnostic methods;
  • (v) Methods of treatment; and
  • (vi) Kits.
  • 5.1 Definitions
  • As used herein, the term “about” or “approximately” means within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, i.e., the limitations of the measurement system. For example, “about” can mean within 3 or more than 3 standard deviations, per the practice in the art. Alternatively, “about” can mean a range of up to 20%, preferably up to 10%, more preferably up to 5%, and more preferably still up to 1% of a given value. Alternatively, particularly with respect to biological systems or processes, the term can mean within an order of magnitude, preferably within 5-fold, and more preferably within 2-fold, of a value.
  • As used herein, the term “antibody” means not only intact antibody molecules, but also fragments of antibody molecules that retain immunogen-binding ability. Such fragments are also well known in the art and are regularly employed both in vitro and in vivo. Accordingly, as used herein, the term “antibody” means not only intact immunoglobulin molecules but also the well-known active fragments F(ab′)2, and Fab. F(ab′)2, and Fab fragments that lack the Fc fragment of intact antibody, clear more rapidly from the circulation, and may have less non-specific tissue binding of an intact antibody (Wahl et al., J. Nucl. Med. 24:316-325 (1983). The antibodies of the invention comprise whole native antibodies, bispecific antibodies; chimeric antibodies; Fab, Fab′, single chain V region fragments (scFv), fusion polypeptides, humanized antibodies derived from a non-human antibody, and unconventional antibodies.
  • As used herein, the term “vector” refers to any genetic element, such as a plasmid, phage, transposon, cosmid, chromosome, virus, virion, etc., which is capable of replication when associated with the proper control elements and which can transfer gene sequences into cells. Thus, the term includes cloning and expression vehicles, as well as viral vectors and plasmid vectors.
  • As used herein, the term “expression vector” refers to a recombinant nucleic acid sequence, e.g., a recombinant DNA molecule, containing a desired coding sequence and appropriate nucleic acid sequences necessary for the expression of the operably linked coding sequence in a particular host organism. Nucleic acid sequences necessary for expression in prokaryotes usually include a promoter, an operator (optional), and a ribosome binding site, often along with other sequences. Eukaryotic cells are known to utilize promoters, enhancers, and termination and polyadenylation signals.
  • As used herein, the term “treating” or “treatment” refers to clinical intervention in an attempt to alter the disease course of the individual or cell being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Therapeutic effects of treatment include, without limitation, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastases, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis. By preventing progression of a disease or disorder, a treatment can prevent deterioration due to a disorder in an affected or diagnosed subject or a subject suspected of having the disorder, but also a treatment may prevent the onset of the disorder or a symptom of the disorder in a subject at risk for the disorder or suspected of having the disorder.
  • An “effective amount” (or “therapeutically effective amount”) is an amount sufficient to affect a beneficial or desired clinical result upon treatment. An effective amount can be administered to a subject in one or more doses. In terms of treatment, an effective amount is an amount that is sufficient to palliate, ameliorate, stabilize, reverse or slow the progression of the disease (e.g., a cancer), or otherwise reduce the pathological consequences of the disease (e.g., a cancer). The effective amount is generally determined by the physician on a case-by-case basis and is within the skill of one in the art. Several factors are typically taken into account when determining an appropriate dosage to achieve an effective amount. These factors include age, sex and weight of the subject, the condition being treated, the severity of the condition and the form and effective concentration of the immunoresponsive cells administered.
  • As used herein, the term “subject” refers to any animal (e.g., a mammal), including, but not limited to, humans, non-human primates, rodents, and the like (e.g., which is to be the recipient of a particular treatment, or from whom cells are harvested).
  • As used herein, the term “an anti-cancer effect” means one or more of a reduction in aggregate cancer cell mass, a reduction in cancer cell growth rate, a reduction in cancer cell proliferation, a reduction in tumor mass, a reduction in tumor volume, a reduction in tumor cell proliferation, a reduction in tumor growth rate, a reduction in tumor metastasis and/or an increase in the proportion of senescent cancer cells.
  • 5.2 TALKs
  • The human anaplastic lymphoma kinase (ALK) gene is located at chr 2p23, contains 29 exons, and encodes a 1,620 amino acid, 220 kDa classical insulin superfamily tyrosine kinase. The mature human ALK protein undergoes post-translational N-linked glycosylation and consists of an extracellular ligand-binding domain, a transmembrane domain, and a single intracellular tyrosine kinase domain.
  • The term “TALK” may be used to refer to the truncated isoforms of ALK referred to herein and the mRNA and corresponding cDNA molecule encoding them.
  • In certain non-limiting embodiments, the present invention provides for an ALK isoform comprising exons 20-29 but lacking the transmembrane domain and/or extracellular domain. The TALK arises as a result of the establishment of a de novo alternative transcriptional initiation (“ATI”) site rather than a genomic rearrangement or a genomic aberration at the ALK locus. Thus, the TALK is termed as ALKATI.
  • In certain non-limiting embodiments, the ALK isoform is not comprised in fusion with a portion of another native protein, for example arising from a translocation event, for example, where the 3′ portion of ALK is fused to a portion of the ATIC, C2orf44, CARS, CLTC, EML4, FN1, KIF5B, KLC1, MSN, NPM1, PPFIBP1, RANBP2, SEC31A, SQSTM1, STRN, TFG, TPM3, TPM4, or VCL protein (see the COMIC database, http://cancer.sanger.ac.uk/cancergenome/projects/cosmic/)
  • In certain non-limiting embodiments, the present invention provides for an ALK isoform encoded by a mRNA transcript comprising exons 20-29 and a portion but not all of intron 19, but not exons comprised in the transmembrane domain and/or extracellular domain of ALK. In certain non-limiting embodiments, the present invention provides for an ALK isoform encoded by a mRNA transcript comprising exons 20-29 and a portion but not all of intron 19, but not exons 1-19, of the ALK gene, either individually or in any combination. In certain non-limiting embodiments, the present invention provides for a nucleic acid molecule comprising the transcript. In certain non-limiting embodiments, the size of the transcript is about 2300-2600 bases, or about 2500 bases, or about 2513 bases. Certain non-limiting embodiments of the transcript comprise about 400 bases of intron 19 upstream of exon 20. Certain non-limiting embodiments of the invention provide for a cDNA molecule corresponding to said mRNA transcript. Said cDNA molecule may optionally be operably linked to a promoter and/or incorporated into a nucleic acid vector. Said promoter may be positioned directly before the coding sequence. In certain non-limiting embodiments, the promoter is a heterologous promoter (that is to say, not the ALK promoter). In certain non-limiting embodiments, said mRNA transcript or cDNA molecule lacks nucleic acid sequence encoding at least a 10 or 20 amino acid long fragment of a protein other than ALK.
  • In certain non-limiting embodiments, the ALK is human ALK.
  • In certain non-limiting embodiments, the corresponding human ALK gene has a sequence as provided by NCBI Reference Sequence: NG_009445.1 Accession No. NG_009445 or an allelic variant thereof.
  • In certain non-limiting embodiments, the human ALK protein has the amino acid sequence provided by NCBI Reference Sequence: NP_004295.2 Accession No. NP_004295 (provided below) or an allelic variant thereof.
  • [SEQ ID NO: 1]
       1 mgaigllwll plllstaavg sgmgtgqrag spaagpplqp replsysrlq rkslavdfvv
      61 pslfrvyard lllppsssel kagrpeargs laldcapllr llgpapgvsw tagspapaea
     121 rtlsrvlkgg svrklrrakq lvlelgeeai legcvgppge aavgllqfnl selfswwirq
     181 gegrlrirlm pekkasevgr egrlsaaira sqprllfqif gtghsslesp tnmpspspdy
     241 ftwnltwimk dsfpflshrs ryglecsfdf pceleysppl hdlrnqswsw rripseeasq
     301 mdlldgpgae rskemprgsf lllntsadsk htilspwmrs ssehctlavs vhrhlqpsgr
     361 yiaqllphne aareillmpt pgkhgwtvlq grigrpdnpf rvaleyissg nrslsavdff
     421 alkncsegts pgskmalqss ftcwngtvlq lgqacdfhqd caqgedesqm crklpvgfyc
     481 nfedgfcgwt qgtlsphtpq wqvrtlkdar fqdhqdhall lsttdvpase satvtsatfp
     541 apiksspcel rmswlirgvl rgnvslvlve nktgkeqgrm vwhvaayegl slwqwmvlpl
     601 ldvsdrfwlq mvawwgqgsr aivafdnisi sldcyltisg edkilqntap ksrnlfernp
     661 nkelkpgens prqtpifdpt vhwlfttcga sgphgptqaq cnnayqnsnl svevgsegpl
     721 kgiqiwkvpa tdtysisgyg aaggkggknt mmrshgvsvl gifnlekddm lyilvgqqge
     781 dacpstnqli qkvcigennv ieeeirvnrs vhewaggggg gggatyvfkm kdgvpvplii
     841 aaggggrayg aktdtfhper lennssvlgl ngnsgaaggg ggwndntsll wagkslqega
     901 tgghscpqam kkwgwetrgg fggggggcss ggggggyigg naasnndpem dgedgvsfis
     961 plgilytpal kvmeghgevn ikhylncshc evdechmdpe shkvicfcdh gtvlaedgvs
    1021 civsptpeph lplslilsvv tsalvaalvl afsgimivyr rkhqelqamq melqspeykl
    1081 sklrtstimt dynpnycfag ktssisdlke vprknitlir glghgafgev yegqvsgmpn
    1141 dpsplqvavk tlpevcseqd eldflmeali iskfnhqniv rcigvslqsl prfillelma
    1201 ggdlksflre trprpsqpss lamldllhva rdiacgcqyl eenhfihrdi aarnclltcp
    1261 gpgrvakigd fgmardiyra syyrkggcam lpvkwmppea fmegiftskt dtwsfgvllw
    1321 eifslgympy psksnqevle fvtsggrmdp pkncpgpvyr imtqcwqhqp edrpnfaiil
    1381 erieyctqdp dvintalpie ygplveeeek vpvrpkdpeg vppllvsqqa kreeerspaa
    1441 ppplpttssg kaakkptaae isvrvprgpa vegghvnmaf sqsnppselh kvhgsrnkpt
    1501 slwnptygsw ftekptkknn piakkephdr gnlglegsct vppnvatgrl pgasllleps
  • In certain non-limiting embodiments, the untruncated or wild type human ALK mRNA has the nucleotide sequence provided by NCBI Reference Sequence: NM_004304.4 Accession No. NM_004304 (provided below) or an allelic variant thereof.
  • [SEQ ID NO: 2]
       1 agctgcaagt ggcgggcgcc caggcagatg cgatccagcg gctctggggg cggcagcggt
      61 ggtagcagct ggtacctccc gccgcctctg ttcggagggt cgcggggcac cgaggtgctt
     121 tccggccgcc ctctggtcgg ccacccaaag ccgcgggcgc tgatgatggg tgaggagggg
     181 gcggcaagat ttcgggcgcc cctgccctga acgccctcag ctgctgccgc cggggccgct
     241 ccagtgcctg cgaactctga ggagccgagg cgccggtgag agcaaggacg ctgcaaactt
     301 gcgcagcgcg ggggctggga ttcacgccca gaagttcagc aggcagacag tccgaagcct
     361 tcccgcagcg gagagatagc ttgagggtgc gcaagacggc agcctccgcc ctcggttccc
     421 gcccagaccg ggcagaagag cttggaggag ccaaaaggaa cgcaaaaggc ggccaggaca
     481 gcgtgcagca gctgggagcc gccgttctca gccttaaaag ttgcagagat tggaggctgc
     541 cccgagaggg gacagacccc agctccgact gcggggggca ggagaggacg gtacccaact
     601 gccacctccc ttcaaccata gtagttcctc tgtaccgagc gcagcgagct acagacgggg
     661 gcgcggcact cggcgcggag agcgggaggc tcaaggtccc agccagtgag cccagtgtgc
     721 ttgagtgtct ctggactcgc ccctgagctt ccaggtctgt ttcatttaga ctcctgctcg
     781 cctccgtgca gttgggggaa agcaagagac ttgcgcgcac gcacagtcct ctggagatca
     841 ggtggaagga gccgctgggt accaaggact gttcagagcc tcttcccatc tcggggagag
     901 cgaagggtga ggctgggccc ggagagcagt gtaaacggcc tcctccggcg ggatgggagc
     961 catcgggctc ctgtggctcc tgccgctgct gctttccacg gcagctgtgg gctccgggat
    1021 ggggaccggc cagcgcgcgg gctccccagc tgcggggccg ccgctgcagc cccgggagcc
    1081 actcagctac tcgcgcctgc agaggaagag tctggcagtt gacttcgtgg tgccctcgct
    1141 cttccgtgtc tacgcccggg acctactgct gccaccatcc tcctcggagc tgaaggctgg
    1201 caggcccgag gcccgcggct cgctagctct ggactgcgcc ccgctgctca ggttgctggg
    1261 gccggcgccg ggggtctcct ggaccgccgg ttcaccagcc ccggcagagg cccggacgct
    1321 gtccagggtg ctgaagggcg gctccgtgcg caagctccgg cgtgccaagc agttggtgct
    1381 ggagctgggc gaggaggcga tcttggaggg ttgcgtcggg ccccccgggg aggcggctgt
    1441 ggggctgctc cagttcaatc tcagcgagct gttcagttgg tggattcgcc aaggcgaagg
    1501 gcgactgagg atccgcctga tgcccgagaa gaaggcgtcg gaagtgggca gagagggaag
    1561 gctgtccgcg gcaattcgcg cctcccagcc ccgccttctc ttccagatct tcgggactgg
    1621 tcatagctcc ttggaatcac caacaaacat gccttctcct tctcctgatt attttacatg
    1681 gaatctcacc tggataatga aagactcctt ccctttcctg tctcatcgca gccgatatgg
    1741 tctggagtgc agctttgact tcccctgtga gctggagtat tcccctccac tgcatgacct
    1801 caggaaccag agctggtcct ggcgccgcat cccctccgag gaggcctccc agatggactt
    1861 gctggatggg cctggggcag agcgttctaa ggagatgccc agaggctcct ttctccttct
    1921 caacacctca gctgactcca agcacaccat cctgagtccg tggatgagga gcagcagtga
    1981 gcactgcaca ctggccgtct cggtgcacag gcacctgcag ccctctggaa ggtacattgc
    2041 ccagctgctg ccccacaacg aggctgcaag agagatcctc ctgatgccca ctccagggaa
    2101 gcatggttgg acagtgctcc agggaagaat cgggcgtcca gacaacccat ttcgagtggc
    2161 cctggaatac atctccagtg gaaaccgcag cttgtctgca gtggacttct ttgccctgaa
    2221 gaactgcagt gaaggaacat ccccaggctc caagatggcc ctgcagagct ccttcacttg
    2281 ttggaatggg acagtcctcc agcttgggca ggcctgtgac ttccaccagg actgtgccca
    2341 gggagaagat gagagccaga tgtgccggaa actgcctgtg ggtttttact gcaactttga
    2401 agatggcttc tgtggctgga cccaaggcac actgtcaccc cacactcctc aatggcaggt
    2461 caggacccta aaggatgccc ggttccagga ccaccaagac catgctctat tgctcagtac
    2521 cactgatgtc cccgcttctg aaagtgctac agtgaccagt gctacgtttc ctgcaccgat
    2581 caagagctct ccatgtgagc tccgaatgtc ctggctcatt cgtggagtct tgaggggaaa
    2641 cgtgtccttg gtgctagtgg agaacaaaac cgggaaggag caaggcagga tggtctggca
    2701 tgtcgccgcc tatgaaggct tgagcctgtg gcagtggatg gtgttgcctc tcctcgatgt
    2761 gtctgacagg ttctggctgc agatggtcgc atggtgggga caaggatcca gagccatcgt
    2821 ggcttttgac aatatctcca tcagcctgga ctgctacctc accattagcg gagaggacaa
    2881 gatcctgcag aatacagcac ccaaatcaag aaacctgttt gagagaaacc caaacaagga
    2941 gctgaaaccc ggggaaaatt caccaagaca gacccccatc tttgacccta cagttcattg
    3001 gctgttcacc acatgtgggg ccagcgggcc ccatggcccc acccaggcac agtgcaacaa
    3061 cgcctaccag aactccaacc tgagcgtgga ggtggggagc gagggccccc tgaaaggcat
    3121 ccagatctgg aaggtgccag ccaccgacac ctacagcatc tcgggctacg gagctgctgg
    3181 cgggaaaggc gggaagaaca ccatgatgcg gtcccacggc gtgtctgtgc tgggcatctt
    3241 caacctggag aaggatgaca tgctgtacat cctggttggg cagcagggag aggacgcctg
    3301 ccccagtaca aaccagttaa tccagaaagt ctgcattgga gagaacaatg tgatagaaga
    3361 agaaatccgt gtgaacagaa gcgtgcatga gtgggcagga ggcggaggag gagggggtgg
    3421 agccacctac gtatttaaga tgaaggatgg agtgccggtg cccctgatca ttgcagccgg
    3481 aggtggtggc agggcctacg gggccaagac agacacgttc cacccagaga gactggagaa
    3541 taactcctcg gttctagggc taaacggcaa ttccggagcc gcaggtggtg gaggtggctg
    3601 gaatgataac acttccttgc tctgggccgg aaaatctttg caggagggtg ccaccggagg
    3661 acattcctgc ccccaggcca tgaagaagtg ggggtgggag acaagagggg gtttcggagg
    3721 gggtggaggg gggtgctcct caggtggagg aggcggagga tatataggcg gcaatgcagc
    3781 ctcaaacaat gaccccgaaa tggatgggga agatggggtt tccttcatca gtccactggg
    3841 catcctgtac accccagctt taaaagtgat ggaaggccac ggggaagtga atattaagca
    3901 ttatctaaac tgcagtcact gtgaggtaga cgaatgtcac atggaccctg aaagccacaa
    3961 ggtcatctgc ttctgtgacc acgggacggt gctggctgag gatggcgtct cctgcattgt
    4021 gtcacccacc ccggagccac acctgccact ctcgctgatc ctctctgtgg tgacctctgc
    4081 cctcgtggcc gccctggtcc tggctttctc cggcatcatg attgtgtacc gccggaagca
    4141 ccaggagctg caagccatgc agatggagct gcagagccct gagtacaagc tgagcaagct
    4201 ccgcacctcg accatcatga ccgactacaa ccccaactac tgctttgctg gcaagacctc
    4261 ctccatcagt gacctgaagg aggtgccgcg gaaaaacatc accctcattc ggggtctggg
    4321 ccatggcgcc tttggggagg tgtatgaagg ccaggtgtcc ggaatgccca acgacccaag
    4381 ccccctgcaa gtggctgtga agacgctgcc tgaagtgtgc tctgaacagg acgaactgga
    4441 tttcctcatg gaagccctga tcatcagcaa attcaaccac cagaacattg ttcgctgcat
    4501 tggggtgagc ctgcaatccc tgccccggtt catcctgctg gagctcatgg cggggggaga
    4561 cctcaagtcc ttcctccgag agacccgccc tcgcccgagc cagccctcct ccctggccat
    4621 gctggacctt ctgcacgtgg ctcgggacat tgcctgtggc tgtcagtatt tggaggaaaa
    4681 ccacttcatc caccgagaca ttgctgccag aaactgcctc ttgacctgtc caggccctgg
    4741 aagagtggcc aagattggag acttcgggat ggcccgagac atctacaggg cgagctacta
    4801 tagaaaggga ggctgtgcca tgctgccagt taagtggatg cccccagagg ccttcatgga
    4861 aggaatattc acttctaaaa cagacacatg gtcctttgga gtgctgctat gggaaatctt
    4921 ttctcttgga tatatgccat accccagcaa aagcaaccag gaagttctgg agtttgtcac
    4981 cagtggaggc cggatggacc cacccaagaa ctgccctggg cctgtatacc ggataatgac
    5041 tcagtgctgg caacatcagc ctgaagacag gcccaacttt gccatcattt tggagaggat
    5101 tgaatactgc acccaggacc cggatgtaat caacaccgct ttgccgatag aatatggtcc
    5161 acttgtggaa gaggaagaga aagtgcctgt gaggcccaag gaccctgagg gggttcctcc
    5221 tctcctggtc tctcaacagg caaaacggga ggaggagcgc agcccagctg ccccaccacc
    5281 tctgcctacc acctcctctg gcaaggctgc aaagaaaccc acagctgcag agatctctgt
    5341 tcgagtccct agagggccgg ccgtggaagg gggacacgtg aatatggcat tctctcagtc
    5401 caaccctcct tcggagttgc acaaggtcca cggatccaga aacaagccca ccagcttgtg
    5461 gaacccaacg tacggctcct ggtttacaga gaaacccacc aaaaagaata atcctatagc
    5521 aaagaaggag ccacacgaca ggggtaacct ggggctggag ggaagctgta ctgtcccacc
    5581 taacgttgca actgggagac ttccgggggc ctcactgctc ctagagccct cttcgctgac
    5641 tgccaatatg aaggaggtac ctctgttcag gctacgtcac ttcccttgtg ggaatgtcaa
    5701 ttacggctac cagcaacagg gcttgccctt agaagccgct actgcccctg gagctggtca
    5761 ttacgaggat accattctga aaagcaagaa tagcatgaac cagcctgggc cctgagctcg
    5821 gtcgcacact cacttctctt ccttgggatc cctaagaccg tggaggagag agaggcaatg
    5881 gctccttcac aaaccagaga ccaaatgtca cgttttgttt tgtgccaacc tattttgaag
    5941 taccaccaaa aaagctgtat tttgaaaatg ctttagaaag gttttgagca tgggttcatc
    6001 ctattctttc gaaagaagaa aatatcataa aaatgagtga taaatacaag gcccagatgt
    6061 ggttgcataa ggtttttatg catgtttgtt gtatacttcc ttatgcttct ttcaaattgt
    6121 gtgtgctctg cttcaatgta gtcagaatta gctgcttcta tgtttcatag ttggggtcat
    6181 agatgtttcc ttgccttgtt gatgtggaca tgagccattt gaggggagag ggaacggaaa
    6241 taaaggagtt atttgtaatg actaaaa
  • In certain non-limiting embodiments, the present invention provides for an isolated nucleic acid molecule comprising the nucleic acid sequence set forth in SEQ ID NO: 11 as shown in FIG. 6D, or a sequence that is at least 80%, at least 85%, at least 90%, or at least 95% homologous thereto, including sequences that are not identical to SEQ ID NO: 11, but lacking sequence encoding exons 1-19 of ALK, either individually or in any combination.
  • In certain non-limiting embodiments, the nucleic acid molecule comprises a nucleotide sequence encoding the amino acid sequence set forth in SEQ ID NO: 12 as shown in FIG. 6E, or a sequence that is at least 80%, at least 85%, at least 90%, or at least 95% homologous thereto, including sequences that are not identical to SEQ ID NO: 12. In certain non-limiting embodiments, the nucleic acid molecule comprises a nucleotide sequence encoding an amino acid sequence that comprises at least one (e.g., no more than 2, no more than 3, no more than 4, no more than 5, no more than 6, no more than 7, no more than 8, no more than 9, no more than 10, no more than 11, no more than 12, no more than 13, no more than 14, no more than 15, no more than 16, no more than 17, no more than 18, no more than 19, or no more than 20) amino acid variation relative to SEQ ID NO: 12, where the resulting sequence is not found in a naturally occurring protein, e.g., a wild-type protein.
  • In certain non-limiting embodiments, the nucleic acid molecule comprises a nucleotide sequence encoding amino acids 3-552 of SEQ ID NO: 12 or a sequence that is at least 80%, at least 85%, at least 90%, or at least 95% homologous thereto, including sequences that are not identical to amino acids 3-552 of SEQ ID NO: 12. In certain non-limiting embodiments, the nucleic acid molecule comprises a nucleotide sequence encoding an amino acid sequence that comprises at least one (e.g., no more than 2, no more than 3, no more than 4, no more than 5, no more than 6, no more than 7, no more than 8, no more than 9, no more than 10, no more than 11, no more than 12, no more than 13, no more than 14, no more than 15, no more than 16, no more than 17, no more than 18, no more than 19, or no more than 20) amino acid variation relative to amino acids 3-552 of SEQ ID NO: 12, where the resulting sequence is not found in a naturally occurring protein, e.g., a wild-type protein.
  • In certain non-limiting embodiments, the nucleic acid molecule comprises a nucleotide sequence encoding amino acids 21-552 of SEQ ID NO: 12 or a sequence that is at least 80%, at least 85%, at least 90%, or at least 95% homologous thereto, including sequences that are not identical to amino acids 21-552 of SEQ ID NO: 12. In certain non-limiting embodiments, the nucleic acid molecule comprises a nucleotide sequence encoding an amino acid sequence that comprises at least one (e.g., no more than 2, no more than 3, no more than 4, no more than 5, no more than 6, no more than 7, no more than 8, no more than 9, no more than 10, no more than 11, no more than 12, no more than 13, no more than 14, no more than 15, no more than 16, no more than 17, no more than 18, no more than 19, or no more than 20) amino acid variation relative to amino acids 21-552 of SEQ ID NO: 12, where the resulting sequence is not found in a naturally occurring protein, e.g., a wild-type protein.
  • In certain non-limiting embodiments, the amino acid sequence has altered glycosylation patterns compared to a naturally occurring protein, including but not limited to, an amino acid sequence produced in non-human cells, e.g., yeast cells, insect cells, CHO cells, etc.
  • In certain non-limiting embodiments, the present invention provides for an isolated nucleic acid molecule, which is a cDNA molecule, comprises nucleic acids 405-2063 of the nucleic acid sequence set forth in SEQ ID NO: 11 as shown in FIG. 6D or a sequence that is at least 80%, at least 85%, at least 90%, or at least 95% homologous thereto, including sequences that are not identical to nucleic acids 405-2063 of SEQ ID NO: 11.
  • In certain non-limiting embodiments, the present invention provides for an isolated nucleic acid molecule, which is a cDNA molecule, comprises nucleic acids 411-2063 of the nucleic acid sequence set forth in SEQ ID NO: 11 as shown in FIG. 6D or a sequence that is at least 80%, at least 85%, at least 90%, or at least 95% homologous thereto, including sequences that are not identical to nucleic acids 411-2063 of SEQ ID NO: 11.
  • In certain non-limiting embodiments, the present invention provides for an isolated nucleic acid molecule, which is a cDNA molecule, comprises nucleic acids 465-2063 of the nucleic acid sequence set forth in SEQ ID NO: 11 as shown in FIG. 6D or a sequence that is at least 80%, at least 85%, at least 90%, or at least 95% homologous thereto, including sequences that are not identical to nucleic acids 465-2063 of SEQ ID NO: 11.
  • In certain non-limiting embodiments, said nucleic acid molecule is operably linked to a promoter, which may be a heterologous promoter. In certain non-limiting embodiments, the nucleic acid molecule comprises a cDNA molecule. In certain non-limiting embodiments, the isolated nucleic acid molecule is comprised in a vector which may, for example, be an expression vector or a virus.
  • In certain non-limiting embodiments, the present invention provides for a host cell comprising a vector that comprises the nucleic acid molecules described herein.
  • Sequence homology or sequence identity may be measured using sequence analysis software (for example, Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 1710 University Avenue, Madison, Wis. 53705, BLAST, BESTFIT, GAP, or PILEUP/PRETTYBOX programs). Such software matches identical or similar sequences by assigning degrees of homology to various substitutions, deletions, and/or other modifications. In an exemplary approach to determining the degree of identity, a BLAST program may be used, with a probability score between e-3 and e-100 indicating a closely related sequence.
  • In certain non-limiting embodiments, the present invention provides for an isolated polypeptide encoded by any of the nucleic acid molecules described herein, including cDNA molecules.
  • In certain non-limiting embodiments, the present invention provides for an isolated polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 12 as shown in FIG. 6E, but lacking the amino acid sequence set forth in SEQ ID NO: 3 (provided below) or a sequence that is at least 80%, at least 85%, at least 90%, or at least about 95% homologous thereto, including sequences that are not identical to SEQ ID NO: 12.
  • [SEQ ID NO: 3]
    MGAIGLLWLLPLLLSTAAVGSGMGTGQRAGSPAAGPPLQPREPL
    SYSRLQRKSLAVDEVVPSLERVYARDLLLPPSSSELKAGRPEARGSLALD
    CAPLLRLLGPAPGVSWTAGSPAPAEARTLSRVLKGGSVRKLRRAKQLVLE
    LGEEAILEGCVGPPGEAAVGLLQFNLSELFSWWIRQGEGRLRIRLMPEKK
    ASEVGREGRLSAAIRASQPRLLFQIFGTGHSSLESPTNMPSPSPDYFTWN
    LTWIMKDSFPFLSHRSRYGLECSFDFPCELEYSPPLHDLRNQSWSWRRIP
    SEEASQMDLLDGPGAERSKEMPRGSFLLLNTSADSKHTILSPWMRSSSEH
    CTLAVSVHRHLQPSGRYIAQLLPHNEAAREILLMPTPGKHGWTVLQGRIG
    RPDNPFRVALEYISSGNRSLSAVDFFALKNCSEGTSPGSKMALQSSFTCW
    NGTVLQLGQACDFHQDCAQGEDESQMCRKLPVGFYCNFEDGFCGWTQGTL
    SPHTPQWQVRTLKDARFQDHQDHALLLSTTDVPASESATVTSATFPAPIK
    SSPCELRMSWLIRGVLRGNVSLVLVENKTGKEQGRMVWHVAAYEGLSLWQ
    WMVLPLLDVSDREWLQMVAWWGQGSRAIVAFDNISISLDCYLTISGEDKI
    LQNTAPKSRNLFERNPNKELKPGENSPRQTPIFDPTVHWLFTTCGASGPH
    GPTQAQCNNAYQNSNLSVEVGSEGPLKGIQIWKVPATDTYSISGYGAAGG
    KGGKNTMMRSHGVSVLGIFNLEKDDMLYILVGQQGEDACPSTNQLIQKVC
    IGENNVIEEEIRVNRSVHEWAGGGGGGGGATYVFKMKDGVPVPLIIAAGG
    GGRAYGAKTDTFHPERLENNSSVLGLNGNSGAAGGGGGWNDNTSLLWAGK
    SLQEGATGGHSCPQAMKKWGWETRGGFGGGGGGCSSGGGGGGYIGGNAAS
    NNDPEMDGEDGVSFISPLGILYTPALKVMEGHGEVNIKHYLNCSHCEVDE
    CHMDPESHKVICFCDHGTVLAEDGVSCIVSPTPEPHLPLSLILSVVTSAL
    VAALVLAFSGIMIVYRRKHQELQA
  • In certain non-limiting embodiments, the present invention provides for an isolated polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 12 as shown in FIG. 6E, but lacking the amino acid sequence set forth in SEQ ID NO: 4 (provided below) or a sequence that is at least 80%, at least 85%, at least 90%, or at least about 95% homologous thereto, including sequences that are not identical to SEQ ID NO: 12.
  • [SEQ ID NO: 4]
    MGAIGLLWLLPLLLSTAAVGSGMGTGQRAGSPAAGPPLQPREPL
    SYSRLQRKSLAVDFVVPSLFRVYARDLLLPPSSSELKAGRPEARGSLALD
    CAPLLRLLGPAPGVSWTAGSPAPAEARTLSRVLKGGSVRKLRRAKQLVLE
    LGEEAILEGCVGPPGEAAVGLLQFNLSELFSWWIRQGEGRLRIRLMPEKK
    ASEVGREGRLSAAIRASQPRLLFQIFGTGHSSLESPTNMPSPSPDYFTWN
    LTWIMKDSFPFLSHRSRYGLECSFDFPCELEYSPPLHDLRNQSWSWRRIP
    SEEASQMDLLDGPGAERSKEMPRGSFLLLNTSADSKHTILSPWMRSSSEH
    CTLAVSVHRHLQPSGRYIAQLLPHNEAAREILLMPTPGKHGWTVLQGRIG
    RPDNPFRVALEYISSGNRSLSAVDFFALKNCSEGTSPGSKMALQSSFTCW
    NGTVLQLGQACDFHQDCAQGEDESQMCRKLPVGFYCNFEDGFCGWTQGTL
    SPHTPQWQVRTLKDARFQDHQDHALLLSTTDVPASESATVTSATFPAPIK
    SSPCELRMSWLIRGVLRGNVSLVLVENKTGKEQGRMVWHVAAYEGLSLWQ
    WMVLPLLDVSDRFWLQMVAWWGQGSRAIVAFDNISISLDCYLTISGEDKI
    LQNTAPKSRNLFERNPNKELKPGENSPRQTPIFDPTVHWLFTTCGASGPH
    GPTQAQCNNAYQNSNLSVEVGSEGPLKGIQIWKVPATDTYSISGYGAAGG
    KGGKNTMMRSHGVSVLGIFNLEKDDMLYILVGQQGEDACPSTNQLIQKVC
    IGENNVIEEEIRVNRSVHEWAGGGGGGGGATYVFKMKDGVPVPLIIAAGG
    GGRAYGAKTDTFHPERLENNSSVLGLNGNSGAAGGGGGWNDNTSLLWAGK
    SLQEGATGGHSCPQAMKKWGWETRGGFGGGGGGCSSGGGGGGYIGGNAAS
    NNDPEMDGEDGVSFISPLGILYTPALKVMEGHGEVNIKHYLNCSHCEVDE
    CHMDPESHKVICFCDHGTVLAEDGVSCIVSPTPEPHLPLSLILSVVTSA
  • In certain non-limiting embodiments, the polypeptide comprises amino acids 3 to 552 of SEQ ID NO: 12. In certain non-limiting embodiments, the polypeptide comprises amino acids 21 to 552 of SEQ ID NO: 12.
  • In certain non-limiting embodiments, the isolated polypeptide comprises an amino acid sequence that comprises at least one (e.g., no more than 2, no more than 3, no more than 4, no more than 5, no more than 6, no more than 7, no more than 8, no more than 9, no more than 10, no more than 11, no more than 12, no more than 13, no more than 14, no more than 15, no more than 16, no more than 17, no more than 18, no more than 19, or no more than 20) amino acid variation relative to SEQ ID NO: 12, where the resulting sequence is not found in a naturally occurring protein, e.g., a wild-type protein. In certain embodiments, the amino acid sequence comprises at least one amino acid variation relative to SEQ ID NO: 12 but lacks the amino acid sequence set forth in SEQ ID NO: 3. In certain embodiments, the amino acid sequence comprises at least one amino acid variation relative to SEQ ID NO: 12 but lacks the amino acid sequence set forth in SEQ ID NO: 4.
  • In certain non-limiting embodiments, the amino acid sequence has altered glycosylation patterns compared to a naturally occurring protein, including but not limited to, an amino acid sequence produced in non-human cells, e.g., yeast cells, insect cells, CHO cells, etc.
  • Amino acid variations or modifications may include amino acid substitutions, additions and/or deletions. Variations or modifications may be introduced by standard techniques known in the art, such as site-directed mutagenesis and PCR-mediated mutagenesis. In certain embodiments, the at least one amino acid variation comprises at least one amino acid conservative modification. Amino acids can be classified into groups according to their physicochemical properties such as charge and polarity. Conservative amino acid substitutions are ones in which the amino acid residue is replaced with an amino acid within the same group. For example, amino acids can be classified by charge: positively-charged amino acids include lysine, arginine, histidine, negatively-charged amino acids include aspartic acid, glutamic acid, neutral charge amino acids include alanine, asparagine, cysteine, glutamine, glycine, isoleucine, leucine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine. In addition, amino acids can be classified by polarity: polar amino acids include arginine (basic polar), asparagine, aspartic acid (acidic polar), glutamic acid (acidic polar), glutamine, histidine (basic polar), lysine (basic polar), serine, threonine, and tyrosine; non-polar amino acids include alanine, cysteine, glycine, isoleucine, leucine, methionine, phenylalanine, proline, tryptophan, and valine.
  • In certain non-limiting embodiments, the polypeptide comprises amino acids 3 to 552 of SEQ ID NO: 12. In certain non-limiting embodiments, the polypeptide comprises amino acids 21 to 552 of SEQ ID NO: 12.
  • In certain non-limiting embodiments, the present invention provides for an isolated polypeptide comprising the amino acid sequence set forth in residues 1-552 of SEQ ID NO: 12 as shown in FIG. 6E. The polypeptide has an estimated molecular weight of about 61 kDa (e.g., 61.08 kDa).
  • In certain non-limiting embodiments, the present invention provides for an isolated polypeptide comprising the amino acid sequence set forth in residues 3-552 of SEQ ID NO: 12 as shown in FIG. 6E. The polypeptide has an estimated molecular weight of about 61 kDa (e.g., 60.82 kDa).
  • In certain non-limiting embodiments, the present invention provides for an isolated polypeptide comprising the amino acid sequence set forth in residues 21-552 of SEQ ID NO: 12 as shown in FIG. 6E. The polypeptide has an estimated molecular weight of about 59 kDa (e.g., 58.71 kDa).
  • In certain non-limiting embodiments, the polypeptide has an estimated molecular weight of between 58-64, e.g., about 61 kDa, or about 59 kDa.
  • In certain non-limiting embodiments, the polypeptide is linked to a heterologous peptide tag of between about 2-20 amino acids, or between about 3-15 amino acids.
  • In certain non-limiting embodiments, the polypeptide is detectably labeled.
  • In certain non-limiting embodiments, the polypeptide contains at least one conservative amino acid substitution relative to the wild-type sequence.
  • In certain non-limiting embodiments, the present invention provides for an antibody that binds to one of the above-described polypeptides, but that does not bind to a wild-type ALK polyeptide, e.g., one comprising the amino acid sequence set forth in SEQ ID NO: 1.
  • 5.3 Detection of TALKs
  • Detection of TALKs may be performed by any method known in the art. A TALK may be detected in the form of a mRNA transcript, a corresponding cDNA molecule, its encoded polypeptide, or an ATI site in intron 19 of ALK. In one non-limiting embodiment, the TALK is ALKATI.
  • In certain non-limiting embodiments, a mRNA transcript encoding a TALK may be detected by a method comprising probe hybridization (e.g., probe-based NanoString nCounter assay), amplification, reverse transcription, polymerase chain reaction (PCR, e.g., reverse transcription polymerase chain reaction (RT-PCR), quantitative reverse transcriptase PCR, real-time PCR, quantitative real-time PCR), Northern blot, sequencing, microarray, or a combination thereof.
  • In certain non-limiting embodiments, a TALK polypeptide may be detected by a method comprising antibody binding, immunohistochemistry, Western blot, functional (e.g., kinase) assay, enzyme linked immunosorbent assays (ELISAs), radioimmunoassays (RIA), enzyme immunoassays (EIA), mass spectrometry, 1-D or 2-D gel-based analysis systems (e.g., Polyacrylamide gel electrophoresis (PAGE)), immunoprecipitation, or a combination thereof.
  • In certain non-limiting embodiments, a TALK cDNA molecule may be detected by PCR.
  • In certain non-limiting embodiments, an ATI site in intron 19 of an ALK gene may be detected by a method comprising Northern blot, Chromatin immunoprecipitation (ChIP)-seq, ChIP-qPCR, Rapid Amplification of cDNA Ends (RACE)-PCR, or a combination thereof.
  • A detection method may be practiced in a cell or tissue or cell lysate collected from a subject. The cell may be collected as a sample, for example a sample of a tumor or neoplasm or other lesion. The sample may be a peripheral blood sample or other fluid sample (e.g., pleural effusion).
  • In one particular non-limiting embodiment of the invention, the transcriptional start site and 5′-end of a TALK transcript, for example the ALKATI transcript, may be ascertained using the 5′-RACE technique. For example, a tobacco-acid-pyrophosphatase 5′-RACE technique may be performed according to the manufactures protocol (ExactSTART Eukaryotic mRNA RACE Kit, #ES80910, Epicentre) using the following primers: 5′-TCATACACATACGATTTAGGTGACACTATAGAGCGGCCGCCTGCAGGAAA-3′ [SEQ ID NO: 5]; reverse 5′-CAGGTCACTGATGGAGGAGGTCTTGCCAGCAAAGCA-3′ [SEQ ID NO: 6]. The 5′-RACE products may then be sequenced, for example using an Illumina MiSeq System with a 150 bp paired-end protocol according to the manufactures protocol. Alternatively, the following reverse primers may be used: forward primer 5′-CTAATACGACTCACTATAGGGC-3′ [SEQ ID NO: 7], reverse primer 5′-ACACCTGGCCTTCATACACCTCC-3′ [SEQ ID NO: 8].
  • In another particular non-limiting embodiment of the invention, a cDNA molecule of a TALK (e.g., the ALKATI) transcript may be amplified using the ALK specific primers 5′-CACCATCCCATCTCCAGTCTGCTTC-3′ [SEQ ID NO: 9] and 5′-AGAGAAGTGAGTGTGCGACC-3′ [SEQ ID NO: 10].
  • In other particular non-limiting embodiments of the invention, the presence of a TALK may be detected using an ALK-specific antibody as well as a technique that demonstrates a molecular weight lower than wild-type protein, such as but not limited to polyacrylamide gel electrophoresis and Western blot. Non-limiting examples of antibodies that may be used include Anti-α-Tubulin antibody (#T9026-.5ML), Anti-V5 (#MA1-81617), and anti-HA3F10 (#12158167001), Phospho-ALK (Tyr1278/1282/1283) Rabbit mAb (#3983), Phospho-ALK (Tyr1604) Rabbit mAb (#3341), ALK (D5F3) Rabbit mAb (#3633), Phospho-Akt (Ser473) (#4060), Akt (#4685), Phospho-Stat3 (Tyr705) (#9145), Stat3 (#4904), Phospho-S6 (Ser235/236) (#4858), S6 (#2217), Phospho-p44/42 MAPK (Erk1/2) (Thr202/Tyr204) (#4370), p44/42 MAPK (Erk1/2) (#4695), Phospho-MEK1/2 (Ser221) (#2338), MEK1/2 (#9122).
  • In certain non-limiting embodiments, a TALK may be detected via the presence of an ATI site in intron 19 of ALK, for example using ChIP-seq and ChIP-qPCR. The chromatin marks ‘trimethylated histone H3 lysine 4 (H3K4me3)’ and ‘RNApol-II’ are well characterized to enrich at poised and actively transcribed ATI site.17-19
  • In certain non-limiting embodiments, a NanoString nCounter assay with probe sets in ALK exons 1-19, exons 20-29 and intron 19 may be used (see, for FIG. 1H and its legend) to distinguish wild-type ALK (ALKWT), translocated ALK and ALKATI transcripts. NanoString nCounter assay is based on digital detection and direct molecular barcoding of target molecules through the use of a color coded probe pair. The probe pair consists of a Reporter Probe, which carries the signal on its 5′ end, and a Capture Probe which carries a biotin on the 3′ end.51
  • In certain non-limiting embodiments, a TALK may be detected via its cellular localization, in that TALK has a greater presence in the nucleus relative to wild-type ALK. Such studies may be performed, for example, using immunohistochemistry and primary or secondary fluorescent antibody probes.
  • 5.4 Diagnostic Methods
  • The present invention provides for a method of diagnosing a cell as a cancer cell comprising detecting the presence of a TALK in a cell, where the presence of a TALK indicates that the cell is a cancer cell. The cell may be in a sample collected from a subject. In certain non-limiting embodiments, a sample includes, but is not limited to, a clinical sample, cells in culture, cell supernatants, cell lysates, serum, blood plasma, biological fluid (e.g., lymphatic fluid) and tissue samples. The source of the sample may be solid tissue (e.g., from a fresh, frozen, and/or preserved organ, tissue sample, biopsy or aspirate), blood or any blood constituents, bodily fluids (such as, e.g., urine, lymph, cerebral spinal fluid, amniotic fluid, peritoneal fluid or interstitial fluid), or cells from the individual, including circulating tumor cells.
  • In certain non-limiting embodiments, where a TALK is found in a cell, the cell or another cell from the same subject (for example one or more additional cells from the same sample) may be tested for other markers indicative of a diagnosis of cancer, as are known in the art.
  • Non-limiting examples of cancers which may be indicated by the presence of a TALK include melanoma, thyroid carcinoma (e.g., anaplastic thyroid carcinoma), lung adenocarcinoma, lung squamous cell carcinoma, renal clear cell carcinoma, and breast cancer. In one particular non-limiting embodiment, the cancer is melanoma.
  • In certain non-limiting embodiments, the present invention provides a method of determining whether an anti-cancer effect is likely to be produced in a cancer by an ALK inhibitor, comprising determining whether one or more cell of the cancer contains a detectable TALK, wherein the presence of a detectable TALK in the cell indicates that an ALK inhibitor would have an anti-cancer effect on the cancer.
  • In certain non-limiting embodiments, the present invention provides for a method of determining the likelihood that a subject having a cancer may obtain therapeutic benefit from therapy with an ALK inhibitor, comprising determining whether a cancer cell of the subject contains a detectable TALK, where the presence of a detectable TALK indicates that the subject is more likely to benefit from ALK inhibitor therapy than a subject who lacks TALK or an activating genetic mutation of ALK.
  • In certain embodiments, the presence of a detectable TALK comprises the presence of a detectable TALK mRNA transcript, a detectable TALK cDNA molecule corresponding thereto, a detectable TALK polypeptide encoded thereby, and/or a detectable ATI site in intron 19 of an ALK gene in the cell.
  • In certain non-limiting embodiments, a cancer cell of a subject may have a wild-type (or normal) ALK locus and yet express a TALK, and therefore would not be identified as having an activated ALK by genomic testing.
  • 5.5 Methods of Treatment
  • In certain non-limiting embodiments, the invention provides for a method of treating a subject suffering from a cancer, comprising (i) determining the likelihood that the subject would obtain therapeutic benefit from therapy with an ALK inhibitor, comprising determining whether a cancer cell of the subject contains a detectable TALK, where the presence of a detectable TALK indicates that the subject is more likely to benefit from ALK inhibitor therapy than a subject who lacks TALK or an activating genetic mutation of ALK; (ii) where the subject is determined to be more likely to benefit from ALK inhibitor therapy, treating the subject with an ALK inhibitor, and (iii) where the subject is determined to not be more likely to benefit from ALK inhibitor therapy, treating the subject with an alternative therapy which is not an ALK inhibitor but which may be surgical treatment, radiation therapy, or chemotherapy.
  • Non-limiting examples of cancers which may be treated by the above method include melanoma, thyroid carcinoma (e.g., anaplastic thyroid carcinoma), lung adenocarcinoma, lung squamous cell carcinoma, renal clear cell carcinoma, thyroid cancer, and breast cancer. In one particular non-limiting embodiment, the cancer is melanoma.
  • Non-limiting examples of ALK inhibitors which may be used in the above method include crizotinib (“Xalkori®”; (Pfizer); ceritinib (Zykadia®; also known as “LDK-378; Novartis), NVP-TAE684 (Novartis), alectinib (Chugai); AP26113 (Ariad); ASP-3026 (Astellas); CEP-37440 (Teva); NMS-E628 (Nerviano); PF-06463922 (Pfizer); TSR-011 (Tesoro); RXDX-101 (Ignyta Inc.), and X-396 (Xcovery). In one particular non-limiting embodiment, the ALK inhibitor is crizotinib.
  • Subjects treated according to the invention with one or more ALK inhibitor may further be treated with one or more additional cancer treatment, including but not limited to, one or more BRAF inhibitor, one or more MEK inhibitor, one or more immunologic inhibitor (e.g., an anti-CTLA4 antibody, an anti-PD1 antibody, an anti-PD-L1 antibody), one or more CDK4 inhibitor, one or more CDK6 inhibitor, one or more alklyating agent, one or more topoisomerase inhibitor, one or more anti-metabolite, one or more anti-microtubule agent, one or more cytotoxic antibiotic, radiation therapy, chemotherapy, or a combination thereof. In one particular non-limiting embodiment, the subject receives a combination of an anti-CTLA-4 antibody and an PD-1 antibody in addition to an ALK inhibitor. In one non-limiting embodiment, the subject has been treated with a combination of an anti-CTLA-4 antibody and an anti-PD-1 antibody prior to the treatment with an ALK inhibitor. In certain embodiments, the subject further receives radiation and/or chemotherapy.
  • 5.6 Kits
  • In certain non-limiting embodiments, the present invention provides for a kit comprising a means for detecting a TALK as described above, optionally together with written disclosure of one or more method set forth above, and optionally together with a positive control molecule such as a TALK protein and/or nucleic acid.
  • In certain embodiments, the kit comprises means for determining the level of a TALK mRNA transcript, a TALK cDNA molecule corresponding thereto, and/or a TALK polypeptide encoded thereby. The means for determining the level of a TALK mRNA transcript may be probe hybridization (e.g., probe-based NanoString nCounter assay), polymerase chain reaction (PCR, e.g., RT-PCR, quantitative reverse transcriptase PCR, real-time PCR, or quantitative real-time PCR), Northern blot, sequencing, microarray, or a combination thereof.
  • In certain embodiments, the means for determining the level of a TALK cDNA molecule may be PCR.
  • In certain embodiments, the means for determining the level of a TALK polypeptide may be antibody binding, immunohistochemistry, Western blot, a functional (e.g., kinase) assay, enzyme linked immunosorbent assays (ELISAs), radioimmunoassays (RIA), enzyme immunoassays (EIA), mass spectrometry, 1-D or 2-D gel-based analysis systems (e.g., Polyacrylamide gel electrophoresis (PAGE)), immunoprecipitation, or a combination thereof.
  • In certain embodiments, the kit comprises means for determining the presence of an ATI site in intron 19 of an ALK gene in one or more cell of the cancer, which may be Northern blot, Chromatin immunoprecipitation (ChIP)-seq, ChIP-qPCR, Rapid Amplification of cDNA Ends (RACE)-PCR, or a combination thereof.
  • The present invention further provides for a means for determining the presence of a detectable TALK for use in a method of determining whether an anti-cancer effect is likely to be produced in a cancer by an ALK inhibitor, the method characterized by determining whether one or more cell of the cancer contains a detectable TALK, where the presence of a detectable TALK in the cell indicates that an ALK inhibitor would have an anti-cancer effect on the cancer.
  • Such a kit may therefore comprise one or more nucleic acid probe (e.g., that hybridizes with a TALK mRNA transcript (including a color coded probe pair used in a NanoString nCounter assay), or that hybridizes with a TALK cDNA molecule), primer, and/or primer pair (e.g., that hybridizes with a TALK mRNA transcript or a TALK cDNA molecule), and/or one or more antibody, antibody fragment, or single chain antibody (e.g., that binds specifically to a TALK polypeptide), any of which are optionally directly labeled with a chemical, fluorescent, enzymatic or radioactive marker. Alternatively, detection may be achieved via a secondary probe, enzyme, substrate, ligand, antibody, antibody fragment, single chain antibody, etc., using techniques known in the art.
  • In a specific non-limiting embodiment, the kit comprises the primer pair: 5′-TCATACACATACGATTTAGGTGACACTATAGAGCGGCCGCCTGCAGGAAA-3′ [SEQ ID NO: 5]; reverse 5′-CAGGTCACTGATGGAGGAGGTCTTGCCAGCAAAGCA-3′ [SEQ ID NO: 6], for example, but not by way of limitation, for use in a 5′-RACE technique.
  • In a specific non-limiting embodiment, the kit comprises the primer pair: forward primer 5′-CTAATACGACTCACTATAGGGC-3′ [SEQ ID NO: 7], reverse primer 5′-ACACCTGGCCTTCATACACCTCC-3′ [SEQ ID NO: 8], for example, but not by way of limitation, for use in a 5′-RACE technique.
  • In a specific non-limiting embodiment, the kit comprises the primer pair: 5′-CACCATCCCATCTCCAGTCTGCTTC-3′ [SEQ ID NO: 9] and 5′-AGAGAAGTGAGTGTGCGACC-3′ [SEQ ID NO: 10].
  • In a specific, non-limiting embodiment, the kit comprises an antibody or a fragment thereof that binds to ALK in a region comprised in exons 20-29 and optionally an antibody or a fragment thereof that binds to ALK in a region comprised in exons 1-19.
  • In a specific, non-limiting embodiment, the kit comprises means to identify an ATI site in intron 19 of ALK.
  • In a specific non-limiting embodiment, the kit comprises means to detect nuclear localization of TALK, for example a nucleic acid or antibody probe and a second nucleic acid or antibody probe that specifically binds to a binding partner in the nucleus or a chemical means of identifying or staining the nucleus.
  • 6. EXAMPLE: ALTERNATIVE TRANSCRIPTIONAL INITIATION LEADS TO EXPRESSION OF A NOVEL ALK ISOFORM IN CANCER
  • 6.1 Materials and Methods
  • Human Tumor Samples.
  • The study was approved by the Institutional Review Boards/Ethics Committees of Memorial Sloan-Kettering Cancer Center, New York, and was conducted according to the Declaration of Helsinki. Representative parts of excised tumors were snap frozen in liquid nitrogen or fixed in 4% neutral buffered formalin, embedded in paraffin, processed using routine histologic methods and stained with hematoxylin-eosin. Specimens with insufficient tissue amount or severely degraded nucleic acids were excluded.
  • RNA Sequencing.
  • Total RNA was extracted from fresh-frozen tissue sections (17 metastatic melanoma and 6 thyroid carcinoma) using Qiagen's RNeasy Mini Kit (#74104, Qiagen). The isolated RNA was processed using the TruSeq RNA sample Prep kit (#15026495, Illumina) according to the manufactures protocol. Briefly, the RNA was poly-a selected, reverse transcribed and the obtained cDNA underwent an end-repair process, A-tailing, ligation of the indexes & adapters, and PCR enrichment. The created libraries were sequenced on an Illumina HiSeq-2500 platform with 50, 75 or 100 bp paired-end reads to obtain on average 40 to 100 million reads per sample. Sequencing data were mapped to the human reference genome (hg19) using Bowtie or BWA and analyzed using publicly available software packages: SAMtools26, Tophat27, FusionSeq28, GATK29, Picard (http://picard.sourceforge.net) and IGV30
  • Screening for Aberrantly Expressed Kinases.
  • For initial screening of RNA-seq data, candidate receptor tyrosine kinase genes (RTK) were defined by Gene Ontology annotation GO:0004714 as found in AmiGO31. DEXSeq9 was used to calculate exon level counts using RTK Ensembl Gene IDs. For each gene in each sample, the ratio of reads in the first half of the gene to the second half was calculated. ALK was identified as the top hit.
  • Analysis of Public Datasets.
  • RNA-seq data was downloaded from the Broad Institute GTEx Genotype-Tissue Expression Portal (http://www.broadinstitute.org/gtex/) 2013_09_23 run using exon_quantification data from illuminahiseqrnaseqv2 unc_udu. ALKATI candidates were identified as samples with an ALK expression level of RSEM≥100, ≥500 total reads across all ALK exons, and ≥10× greater average expression (by exon-level RPKM) in exons 20-29 compared to exons 1-19. To confirm ALKATI expression, candidates were manually examined in IGV30. ENCODE ChIP-seq data for H3K27ac, mapped to hg19 and converted to bigwig track format, was downloaded from http://genome.ucsc.edu/ENCODE/dataMatrix/encodeChipMatrixHuman.html.
  • Promoter/Motif Analysis.
  • The proximal cis-regulatory region, chr2:29,445,000-29,447,100, was scanned for transcription factor motifs using FIMO48 with default parameters against the known vertebrate transcription factor motifs in the JASPAR database52.
  • 5′-Rapid Amplification of cDNA Ends (5′-RACE).
  • Two independent 5′-RACE techniques were used to map the ATI site and the 5′-end of the ALKATI transcript. A tobacco-acid-pyrophosphatase 5′-RACE technique according to the manufacture's protocol (ExactSTART Eukaryotic mRNA RACE Kit, #ES80910, Epicentre) using the following primers: 5′-TCATACACATACGATTTAGGTGACACTATAGAGCGGCCGCCTGCAGGAAA-3′ [SEQ ID NO: 5]; reverse 5′-CAGGTCACTGATGGAGGAGGTCTTGCCAGCAAAGCA-3′ [SEQ ID NO: 6]. The 5′-RACE products were sequenced on an Illumina MiSeq System with a 150 bp paired-end protocol according to the manufacture's protocol. The sequencing reads were mapped to the human reference genome (hg19) using BWA, analyzed using Tophat27 and visualized using IGV30. The continuous transcription starting in ALK intron 19 was confirmed with an independent oligonucleotide-based 5′-RACE kit (SMARTer™ RACE cDNA Amplification Kit, #634923, Clontech) according to the manufacture's protocol using the following reverse primers: forward primer 5′-CTAATACGACTCACTATAGGGC-3′ [SEQ ID NO: 7], reverse primer 5′-ACACCTGGCCTTCATACACCTCC-3′ [SEQ ID NO: 8]. The RACE cDNA products were cloned into plasmids (Zero Blunt® TOPO® PCR Cloning Kit, #K2800-20, Invitrogen) and were analyzed with Sanger sequencing using standard procedures. Two lung cancer cell lines (H3122, H2228) with EML4-ALK translocations were used as controls, and as expected, both controls showed the EML4 gene next to ALK exon 20.
  • Chromatin Immunoprecipitation (ChIP)-Seq and ChIP-qPCR.
  • Chromatin was isolated from human tumor tissue and cell lines. Fresh-frozen human tumor tissue (MM-15, MM-74, ATC-28) was sectioned with a microtome and cross-linked in 1% paraformaldehyde for 15 min. The cross-linked tissue samples were quenched in 125 mM Glycine for 10 min, washed in PBS, re-suspended in lysis buffer, and dounced in a Tenbroeck-style tissue grinder and conicated.32 Chromatin isolation from the cell-lines H3122 and SKMEL-524 cells and immunoprecipitation was performed as previously described32. Solubilized chromatin from human tumors and cell lines was immune-precipitated with antibodies against H3K4me3 (#39159, Active Motif), H3K27ac (#Ab4729, Abcam), and RNA-pol II (#39097, Active Motif).
  • ChIP-seq was performed on an Illumina HiSeq2500 with 51-bp single reads. Reads were aligned to the human genome hg19 using the Bowtie alignment software within the Illumina Analysis Pipeline. Duplicate reads were eliminated for subsequent analysis. Peak calling was performed using MACS 1.4 comparing immune-precipitated chromatin with input chromatin.33 ChIP-qPCR was performed on a ViiA™ 7 Real Time PCR System (Life Technologies) using Power SYBR Master Mix (#4367659, Life Technology). ChIP-qPCR primers were designed using primer blast (http://www.ncbi.nlm.nih.gov/tools/primer-blast) and are described in FIGS. 7A and 7B.
  • Ultra-Deep Targeted Sequencing of the Entire ALK Locus.
  • Targeted sequencing of the entire ALK locus was performed using custom hybridization capture probes tiling hg19 chr2:29400000-30300000 (Roche/NimbleGen's SeqCap EZ). This region encompassed the entire genomic footprint of ALK as well as ˜150 kb of upstream sequence. After the genomic DNA was fragmented (E220, Covaris), barcoded sequence libraries (New England Biolabs, Kapa Biosystems) were prepared, and hybridization capture on barcoded pools was performed using custom probes (Nimblegen SeqCap). 250 ng of genomic DNA was used for library construction from 7 separate samples: 2 melanoma tumors (MM-15, MM-74), 1 anaplastic thyroid carcinoma (ATC-28), 2 lung cancer cell lines (H3122, H2228) with EML4-ALK translocations, 1 melanoma (SKMEL-28) and 1 control pool of 10 “normal” blood samples. Libraries were pooled at equimolar concentrations (100 ng per library) and used in the capture reaction as previously described.34 To prevent off-target hybridization, we spiked in a pool of blocker oligonucleotides complementary to the full sequences of all barcoded adaptors. The captured libraries were sequenced on an Illumina HiSeq 2500 to generate 75-bp paired-end reads. Sequence data were de-multiplexed using CASAVA, and aligned to the reference human genome (hg19) using BWA.35
  • Local realignment and quality score recalibration were performed using the Genome Analysis Toolkit (GATK) according to GATK best practices.36 A mean unique target sequence coverage of 1778× per sample (range: 1293×-2188×) was achieved. Sequence data were analyzed to identify single nucleotide variants, small insertions/deletions (indels), and structural rearrangements. Single nucleotide variants were called using muTect37 and were compared to the negative control pool (pooled “normal” blood samples). Variants were retained if the variant allele frequency in the tumor was >5 times than in the negative control and the frequency in the negative control was <0.02. Validated SNPs in the dbSNP database were filtered out. Indels were called using the SomaticIndelDetector tool in GATK36 and were retained if the tumor harbored >3 supporting reads and the frequency in the negative control was <0.02. DELLY was used to search for structural rearrangements.38
  • Bisulfite Sequencing of the Entire ALK Locus.
  • Custom capture of the entire ALK locus was performed using custom hybridisation capture probes tiling the entire genomic footprint of ALK (900 kb, chr2:29400000-30300000) followed by bisulfite sequencing. After fragmentation (E220, Covaris) of 3 g genomic DNA of each sample (MM-15, ATC-28, H3122 and SKMEL-28), libraries were prepared with the KAPA Hyper Prep Kit (#KR0961, Kapa Biosystems) without PCR amplification to preserve the methylation status. 1 μg of each barcoded library was pooled at equimolar concentrations and captured according to the manufacturer's protocol (Roche/NimbleGen's SeqCap EZ). After washing the Dynabeads M-270 (#65306, Life Technologies), the non-biotinylated tumor/cell line DNA was dissociated from the biotinylated capture beads with 0.5M NaOH. The single stranded eluted DNA was used for bisulfite conversion using the EZ DNA Methylation-Gold™ Kit (#D5005, Zymo Research) according to the manufacturer's protocol, except for the 98° C. denaturation step. After bisulfite conversion, the KAPA HiFi Uracil PCR polymerase (#KK280, Kapa Biosystems) was used to amplify the library, the reaction with Agencourt AMPure XP beads (A63881, Beckman Coulter) was purified, and the library was sequenced on an Illumina MiSeq with a 150 bp paired-end protocol according to the manufacturer's instructions. Sequence data were aligned to hg19 and analyzed using BISMARK53. The methylation level at CpG sites was compared across all samples; no methylation was detected in the CHG and CHH contexts. Methylation was first computed as the number of methylated CpG reads vs. the number of total reads covering each CpG site (sites with <10 reads were excluded). A sliding window was used to determine the mean methylation level for every 250 bp region (with at least three CpGs) near the ALK promoter region (chr2:29,444,000-29,452,000). Differential methylation was evaluated using a Mann-Whitney test.
  • Whole-Genome Sequencing.
  • Whole-genome sequencing was performed at the New York Genome Center (New York). Briefly, genomic DNA libraries were prepared from MM-15 and ATC-28 (no matched normal DNA was available) using the Illumina PCR-free kit. Libraries were sequenced on a HiSeq 2500 using Illumina's 100 bp paired-end whole-genome sequencing protocol. Sequence reads were mapped using BWA35 and processed using GATK36. Genome-wide analyses of mutations (HaplotypeCaller36), copy number alterations (FREEC)54, and structural variations (CREST)55 were performed. Mutations were annotated with the Ensembl Variant Effect Predictor56 and filtered to 13 remove common polymorphisms. Non-synonymous mutations along with copy number alterations and structural variations were visualised using Circos57.
  • Array CGH.
  • Genomic DNA samples were labeled using a Bioprime Array CGH Genomic Labeling Kit (#18095-011, Life Technologies) according to the manufacturer's instructions. Briefly, 1 μg tumor DNA and reference DNA (#G1471, Promega) were differentially labeled with dCTP-Cy5 (#45-001-291, GE Healthcare), and dCTP-Cy3 (#45-001-290, GE Healthcare). Genome-wide analysis of DNA copy number changes was conducted using an oligonucleotide SurePrint G3 Human CGH Microarray (#G4447A, Agilent) containing 1 million probes according to the manufacturer's protocol. Slides were scanned using Agilent's microarray scanner G2505B and analyzed using Agilent Genomic Workbench.
  • Interphase Fluorescence In Situ Hybridization (FISH).
  • A commercially available ALK break-apart probe (#06N38-020, Abbott) was used according to the manufacturer's protocol. The probes were hybridized on 5 μm-thick tissue sections. The number and localization of the hybridization signals was assessed in a minimum of 100 interphase nuclei with well-delineated contours. At least 10% of neoplastic cells had to show a split signal to report an ALK rearrangement.
  • Northern Blot.
  • Total RNA was extracted from fresh-frozen tissue or cell lines using Qiagen's RNeasy Mini Kit (#74104, Qiagen). Up to 10 μg RNA was used for running formaldehyde-based Northern analysis according to the manufacturer's protocol using the RNA Ambion NorthernMax® Kit (#AM1940, Ambion). After hybridisation with a 32P labelled probe, consisting of ALK exon 20-29, the membrane was washed and visualised.
  • NanoString.
  • Details of the nCounter Analysis System (NanoString Technologies) were reported previously.39 In brief, two sequence-specific probes were constructed for ALK exons 1-19, intron 19, and exons 20-29, respectively. Four control genes (RPS13, RPL27, RPS20, ACTB) were used for normalization. The probes were complementary to a 100 base pair region of the target mRNA and are listed in FIG. 1H. 100 ng of total RNA from each sample was hybridized, the raw data were normalized to the standard curve generated via the nCounter system, and the average value of the two probes in each target region (exons 1-19, intron 19, exons 20-29) were printed in bar charts using GraphPad Prism software 6.0.
  • Cell Lines.
  • NIH-3T3 mouse embryonic fibroblast cells were obtained from the ‘American Type Culture Collection’ (#CRL-1658, ATCC) and were maintained in DMEM. The Interleukin 3 (IL-3) dependent murine pro B cell line, Ba/F3, was obtained from ‘The Deutsche Sammlung von Mikroorganismen und Zellkulturen’ (#ACC-300, DSMZ) and was cultured in RPMI supplemented with Interleukin 3 (1 ng ml; #403-ML-010, R&D Systems). Melan-a cells were provided by Dr. Dorothy Bennett (St. George's Hospital, University of London, London, UK)40 and were maintained in RPMI supplemented with 200 nM of 12-O-tetradecanoylphorbol-13-acetate (TPA; #4174, Cell Signaling). For retrovirus production, 293T cells (#631507, Clontech) were purchased and cultured in DMEM. All cell culture media contained 10% FBS, L-glutamine (2 mM), penicillin (100 U ml-1), and streptomycin (100 μg ml-1). All cells were cultured at 37° C. in 5% CO2.
  • Plasmids.
  • To investigate the functional roles and the activation of oncogenic signaling pathways, ALKATI, EML4-ALK and ALKF1174L expression constructs were generated. For the ALKATI vector, RNA from MM-15 was reverse-transcribed with anchored oligo(dT) primers into cDNA (#04379012001, Roche), PCR amplified with ALK specific primers 5′-CACCATCCCATCTCCAGTCTGCTTC-3′ [SEQ ID NO: 9] and 5′-AGAGAAGTGAGTGTGCGACC-3′ [SEQ ID NO: 10], and the PCR product was cloned into a pENTR vector (#K2400-20, Life Technologies). The full-length ALK plasmid (HsCD00079531) was purchased from the DF/HCC DNA Resource Core (http://plasmid.med.harvard.edu), and EML4-ALKv1 was synthesized at GeneArt (Life Technologies). Site-directed mutagenesis was performed using QuikChange (#200523, Agilent): For kinase-dead ALKATI (ALKATI-KD), the lysine in the ATP-binding site of the kinase domain was mutated to methionine (p.K1 150M) in ALKATI, and for ALKF1174L, a p.F1174L mutation was introduced into ALKWT. Plasmids were sub-cloned into a pMIG-w vector41 (#12282, www.addgene.org), resulting in MSCV-ALKvariant-IRES-GFP constructs, which were confirmed by digestion and sequencing. An empty pMIG-w vector with green-fluorescent-protein (GFP) was used as control for all ALK expression experiments. To confirm the start codons, the three start codons were mutated from ATG to AAG. For co-immuno-precipitation, ALKATL was cloned into pcDNA3.1/nV5-Dest (#12290-010, Life Technologies) and MSCV N-HA FLAG-Dest (#41033, Addgene). For bioluminescence imaging, a triple modality retroviral reporter plasmid (red fluorescent protein (RFP)—thymidine kinase—luciferase)42 was used.
  • Stable Gene Expression.
  • Retrovirus were produced in 293T cells by standard methods using ecotropic or amphotropic packaging vector and XtremeGene 9 (#06365809001, Roche). We harvested the virus-containing supernatant were harvested for 48, 64, 64 h and 72 hours after transfection. The supernatant was pooled, filtered through a 0.45 μm PVDF membrane, and used for transduction in the presence of polybrene (8 μg ml-1). Transduced stably expressing eGFP+ or RFP+ cells were sorted with a FACSAria II (BD Biosciences).
  • Co-Immunoprecipitation.
  • V5-ALKATI and HA-ALKATI was transiently transfected into 293T cells and after 24 hours, cells were lysed in 10 mM Tris HCl pH 7.5, 1% Triton X-100, 150 mM NaCl, 1 mM EDTA, 1 mM DTT, 1 mM PMSF, proteinase/phosphatase inhibitors. After incubation and centrifugation, 100 μl supernatant was used as input, and 300 μl for immunoprecipitation using the following antibodies: 2 g of anti-V5 antibody (#MA1-81617, Thermo Scientific), 10 μl of EZview Red Anti-HA Affinity Gel (#E6779-1ML, Sigma), 2 g of anti-mouse IgG (#sc-2025, Santa Cruz). 20 μl of Protein A/G UltraLink Resin (#53133, Thermo Scientific) were used for immunoprecipitation. The immune-precipitated material was eluted in 4×SDS loading buffer for immunoblotting.
  • In Vitro Kinase Assay.
  • Stably transduced NIH-3T3 cells were grown in a 15 cm dish, washed in PBS, and lysed in 20 mM Tris pH 8.0, 1% NP-40, 125 mM NaCl, 2.5 mM MgCl2, 1 mM EDTA with proteinase/phosphatase inhibitor. Lysates were incubated on ice, centrifuged, pre-cleared with 25 μl Protein A/G UltraLink Resin (#53133, Thermo Scientific) for 30 min at 4° C. under rotation, and immunoprecipitated with 10 μl ALK (D5F3) XP® Rabbit mAb (#3633) and 25 μl Protein A/G UltraLink Resin. After rotation for 120 min at 4° C., the immunoprecipitated material was washed and used according to the instructions of Universal Tyrosine Kinase Assay Kit (#MK410, Clontech). After the enzymatic reaction, the immunoprecipitated material was mixed with 4×SDS loading buffer for immunoblotting.
  • Immunohistochemistry.
  • Immunohistochemistry was performed on archival FFPE tumor specimens using a standard multimer/DAB detection protocol on a Discovery Ultra system (Roche/Ventana Medical Systems) with appropriate negative and positive controls. The following antibodies were purchased from Cell Signaling and were diluted in SignalStain Antibody Diluent (#8112, Cell Signaling) as indicated: ALK (D5F3) XP® Rabbit mAb (#3633) 1:250, Phospho-Akt (Ser473) (D9E) Rabbit mAb (#4060) 1:50, Phospho-Stat3 (Tyr705) (D3A7) Rabbit mAb (#9145) 1:400, Phospho-S6 Ribosomal Protein (Ser235/236) (D57.2.2E) Rabbit mAb (#4858) 1:400, Phospho-p44/42 MAPK (Erk1/2) (Thr202/Tyr204) (D13.14.4E) Rabbit mAb (#4370) 1:400, cleaved Caspase-3 (Asp175) Antibody (#9661) 1:400. The Anti-Ki67 antibody was purchased from Abcam (#ab15580) and was diluted 1:600.
  • Immunofluorescence.
  • Stably transduced NIH-3T3 cells were grown on coverslips, fixed in 4% formaldehyde, washed in PBS for 15 min at room temperature, washed in PBS for 10 min, and incubated in blocking solution (5% goat serum, 0.1% Triton X-100 in PBS) for 1 hour at room temperature. After aspirating blocking solution, cells were incubated with an ALK (D5F3) Rabbit mAb (#3633, Cell Signaling Technology) diluted 1:1000 in blocking buffer overnight at 4° C. After washing the cells 3× with 0.05% Tween 20 and PBS, cells were incubate with a secondary antibody (#A-11012, Life Technologies) diluted 1:500 in blocking buffer for 2 hours at room temperature. After washing in PBS, slides were mounted with Prolong® Gold Antifade Reagent with DAPI (#8961, Cell Signaling Technology) and imaged with a Leica TCS SP5 II confocal microscope.
  • Immunoblot.
  • Cell lysates were prepared in RIPA buffer (#9806, Cell Signaling) supplemented with Halt protease and phosphatase inhibitor cocktail (#78440, Thermo Scientific). Equal amounts of protein, as measured by BCA protein assay (#23225, Thermo Scientific), were resolved in NuPAGE® Novex® 4-12% Bis-Tris Protein Gels (#NP0321BOX, Life Technologies) and transferred electrophoretically onto a Nitrocellulose 0.45 μm membrane (#162-0115, BioRad). Membranes were blocked for 1 hour at room temperature in 50% Odyssey Blocking Buffer in PBS (#927-40000, LI-COR) and were incubated overnight at 4° C. with the primary antibodies diluted at 1:1000 in 50% Odyssey Blocking Buffer in PBS plus 0.1% Tween 20. Following primary antibodies were used: Anti-α-Tubulin antibody (#T9026-0.5ML, Sigma-Aldrich), Anti-V5 (#MA1-81617, Thermo Scientific), and anti-HA3F10 (#12158167001, Roche); Cell Signaling Technology: Phospho-ALK (Tyr1604) Rabbit mAb (#3341), ALK (D5F3) Rabbit mAb (#3633), Phospho-Akt (Ser473) (D9E) Rabbit mAb (#4060), Akt (pan) (11E7) Rabbit mAb (#4685), Phospho-Stat3 (Tyr705) (D3A7) Rabbit mAb (#9145), Stat3 (79D7) Rabbit mAb (#4904), Phospho-S6 Ribosomal Protein (Ser235/236) (D57.2.2E) Rabbit mAb (#4858), S6 Ribosomal Protein (5G10) Rabbit mAb (#2217), Phospho-p44/42 MAPK (Erk1/2) (Thr202/Tyr204) (D13.14.4E) Rabbit mAb (#4370), p44/42 MAPK (Erk1/2) (137F5) Rabbit mAb (#4695), Phospho-MEK1/2 (Ser221) (166F8) Rabbit mAb (#2338), MEK1/2 Antibody (#9122). After 4 washes of 5 minutes in PBST, membranes were incubated withsecondary antibodies (IRDye 800CW Goat anti-Rabbit #926-32211, 1:20,000, LI-COR; IRDye 680RD Goat anti-Mouse #926-68070, 1:20,000, LI-COR) in 50% Odyssey Blocking Buffer in PBS plus 0,1% Tween 20 for 45 minutes at room temperature. After another 4 washes in PBS-T and a final wash with PBS, membranes were scanned with a LI-COR Odyssey CLx scanner and adjusted using LI-COR Image Studio.
  • Luciferase Reporter Assay.
  • The long terminal repeat in ALK intron 19 at the ATI site (LTR16B2, chr2:29,446,649-29,447,062; 414 bp) was amplified using genomic DNA from patient MM-15 and 5′-GTCCTCATGGCTCAGCTTGT-3′ and 5′-AGCACTACACAGGCCACTTC-3′ primers. The PCR product (chr2:29,446,444-29,447,174; 731 bp) was cloned into pGL4.14-firefly luciferase vector (#E6691, Promega). To determine the promoter activity of LTR16B2, 105 cells were transfected with 500 g pGL4.14-LTR16B2 or vector alone; as internal control, 200 μg pRL-TKRenilla luciferase reporter vector (#E2241, Promega) was co-transfected. Luciferase activity was measured using Dual-Glo Luciferase Assay System (#E2920, Promega) 48 hours after transfection. Promoter activity was calculated by normalising firefly luciferase activity to the control Renilla luciferase activity and compared between pGL4.14-LTR16B2 and vector alone.
  • Flow Cytometry and Fluorescence-Activated Cell Sorting (FACS).
  • Flow cytometry analysis for in vitro transformation assays with Ba/F3 cells was performed on a LSRFortessa (BD Biosciences). GFP- or RFP-positive cells were sorted using a FITC (blue laser) or PE (yellow laser) channel, respectively, on a FACSAria II configured with 5 lasers (BD Biosciences).
  • In Vitro Transformation and Drug Treatment Assays.
  • Ba/F3 cells were stably transduced with MSCV-ALKvariant-IRES-GFP constructs with a multiplicity of infection (MOI) of ˜0.26. Based on the MOI calculations, ˜78% of cells were uninfected, ˜20% of cells were infected with one virus particle, and ˜2% of cells were infected by more than one viral particle. Stably transduced Ba/F3 cells were cultured in RPMI medium supplemented with IL-3 (1 ng ml-1) and the transduction rate of ˜20% was validated using flow cytometry for GFP, that was co-expressed with the ALK variants. For the cell proliferation assay, transduced Ba/F3 cells were transferred into IL-3 depleted RPMI medium and cell growth was quantified every 2-4 days with a luminescence assay (#G7571, Promega). For cell viability assays (ALK inhibitor)-dose response curve, 2000 Ba/F3 cells were plated in quadruplicates in wells of 96-well plates with increasing concentrations of the ALK inhibitors crizotinib (#C-7900, LC laboratories), TAE684 (#CT-TAE684, ChemieTek), or ceritinib (#CT-LDK378, ChemieTek) as indicated. All drugs were suspended in DMSO. The cell viability was assessed after 72 hours by a luminescence assay CellTiter-Glo (#G7571, Promega). Results were normalized to growth of cells in a medium containing an equivalent volume of DMSO. The inhibition curve was determined with GraphPad Prism 6.0 software using the ‘log(inhibitor) vs. response—variable slope’ non-linear regression model. For western blot analysis, 10 million Ba/F3 cells were harvested after 2 hours treatment with crizotinib, washed in ice-cold PBS and lysed in RIPA buffer (#9806, Cell Signaling).
  • In Vivo Tumourigenicity and Drug Treatment Assays.
  • All animal experiments were performed in accordance with a protocol approved by MSKCC Institutional Animal Care and Use Committee. 106 cells stably transfected NIH-3T3 or melan-a cells were re-suspended in 50 l of 1:1 mix of PBS and Matrigel (#356237, BD Biosciences) and the cells were subcutaneously injected into the flanks of 6-8 weeks old female CB17-SCID mice (#CB17SC-F, Taconic). For tumor growth assays, 4 mice were injected with each cell line and 8 tumors were assessed. Tumor sizes were measured with calipers every 2 to 7 days for a period of up to 100 days. For in vivo drug sensitivity studies, 8 mice were injected with the stably transduced NIH-3T3 cells expressing a luciferase reporter construct and the indicated plasmids. When the tumors reached an average size of 200-250 mm3, mice were randomized in a vehicle and or treat group. Mice were orally gavaged once a day with crizotinib (100 mg/kg/d) or vehicle. 8 tumors were measured with calipers every 2 to 3 days and growth curves were visualized with Prism GraphPad 6.0. In parallel, tumor growth was monitored by bioluminescence imaging of anesthetized mice by retro-orbitally injecting d-luciferin (150 mg per kg body weight) and imaging with the IVIS Spectrum Xenogen machine (Caliper Life Sciences). After euthanizing the mice, tumors were explanted either lysed in RIPA buffer (#9806, Cell Signaling Technology), or fixed overnight in 4% paraformaldehyde, washed, embedded in paraffin, and sectioned for hematoxylin-and-eosin (HE) staining or immunohistochemistry.
  • Statistics.
  • All statistical comparisons between two groups were performed by GraphPad Prism software 6.0 using a two-tailed unpaired t test.
  • 6.2 Results and Discussion
  • To identify novel mechanisms of oncogene activation, transcriptome analyses (RNA-seq) of metastatic melanoma and thyroid carcinoma were performed. An algorithm9 was used to investigate the differential expression of exons in receptor tyrosine kinases (RTKs). The analysis was focused on transcripts with a high expression of the kinase domain. A novel ALK transcript in two melanoma (MM-15, MM-74) and in one anaplastic thyroid carcinoma (ATC-28) was identified. The novel transcript contained the ALK exons 20-29 preceded by ˜400 base pairs of intron 19, but not exons 1-19 (FIGS. 1A and 5A). This expression pattern was distinct from ALK wild-type (ALTWT) and ALK translocations. The ALTWT shows expression of all exons, but no expression of introns (FIGS. 1A, 5B), and is commonly found in neuroblastoma in association with activating mutations.10-13 ALK translocations are observed in various cancer types, including lymphoma, sarcoma, and lung cancer and typically occur in intron 19.14-16 Due to preserved splice sites, ALK translocations usually encompass ALK exons 20-29 with little intronic expression (FIGS. 1A and 5C). ALKWT overexpression10, 58-60 and amplification10,60, activating ALK mutations10,11-13, and ALK translocations15,16 are well-established oncogenic drivers in various cancer types, including melanocytic tumors62.
  • To evaluate if the novel ALK transcript arises from alternative transcriptional initiation (ATI), a 5′-rapid amplification of cDNA ends (5′-RACE) was performed. The ATI site was mapped to a 25-bp region in intron 19 and the presence of the novel transcript was confirmed and termed as ALKATI by Northern blot (FIGS. 1B and 6A-6G). ChIPseq and ChIP-qPCR showed that only ALKATI-expressing tumors, but not the controls, had significant enrichment of the chromatin marks trimethylated histone H3 lysine 4 (H3K4me3) and RNA polymerase II at the ATI site, which are characteristic for active promoters17,18 (FIGS. 1C-1E and 7A-7D). Taken together, these data suggest that ALKATI originates from a newly established bona fide ATI site associated with characteristic chromatin alterations.
  • To determine the prevalence of ALKATI expression, more than 5000 samples from 15 different cancer types in the TCGA RNA-seq dataset were screened. ALKATI was expressed in ˜11% of melanoma (38 of 334 tumors) and sporadically in other cancer types, including lung adenocarcinoma, lung squamous cell carcinoma, clear cell renal cell carcinoma, and breast carcinoma (FIGS. 8A-8E). No ALKATI expression was found in more than 1600 samples from 43 different normal tissues in the Genotype-Tissue Expression (GTEx) RNA-seq dataset1, indicating that ALKATI is primarily expressed in cancers, particularly melanoma.
  • To accurately quantify ALKATI expression in clinical specimens, a NanoString nCounter assay with probe sets in ALK exons 1-19, intron 19, and exons 20-29 was developed51 (FIG. 1H). This assay was able to distinguish ALKATI, ALKWT and translocated ALK and identified additional ALKATI-expressing tumors derived from both fresh-frozen and formalin-fixed, paraffin-embedded (FFPE) clinical specimens (FIG. 1F).
  • To determine whether somatic genomic aberrations at the ALK locus contribute to the establishment of the de novo ATI site, comprehensive genetic analyses including interphase fluorescence in situ hybridisation (FISH), genome-wide array-CGH, whole-genome sequencing, and ultra-deep sequencing of the entire ALK locus were performed, results of which are shown in FIGS. 9A-9D and 10A-10F. For example, since transcriptional activation may arise due to genomic rearrangements,20 interphase FISH was performed using probes recognizing the 5′ and 3′ ends of ALK locus, but found no ALK rearrangements (FIGS. 9A and 9B). To examine the ALK locus for previously described small deletions21,22 and tandem duplications,16 genome-wide array-CGH was used, but no genetic alterations was discovered (FIGS. 9C and 9D). Finally, to investigate whether ALKATI could arise through genetic alterations that create a de novo ATI site similar to the recently described TERT promoter mutations in melanoma.23,24 ultra-deep targeted sequencing of the entire ALK locus was performed. No recurrent single nucleotide variations (SNVs), insertions, or deletions were identified in the ALK exons and introns (FIGS. 10A-10D). In summary, no genomic aberrations that could account for the de novo expression of ALKATI were found. Reasoning that local genomic aberrations are usually cis-acting and only affect the expression of the effected allele,25,43 the SNVs in the DNA-, RNA- and ChIP-sequencing data were analyzed. Compared to genomic DNA, similar allelic SNV frequencies in the RNA- and ChIP-sequencing data were found, which indicates that both ALK alleles are actively transcribed and decorated with H3K4me3 (FIG. 1G). The bi-allelic ALKATI expression indicates that the transcriptional activation of ALKATI is independent of genetic aberrations. However, aberrations affecting trans-acting elements, such as transcription factors or chromatin modifiers, may contribute to ALKATI expression.
  • The ALK ATI region contains transposable elements, including a long-terminal repeat (LTR) in intron 19 and a long-interspaced element (LINE) in intron 18, both of which can regulate transcription44,45 (FIG. 11A). To evaluate if CpG methylation of these elements might contribute to ALKATI expression, bisulfite sequencing of the entire ALK locus was performed. Compared to the controls, the ALKATI-expressing samples showed lower CpG methylation in regions flanking the ATI site, including the LINE (FIG. 11B-D). The LTR contained only few CpGs with low methylation levels in all samples. The ENCODE data46 revealed a DNase I hypersensitivity cluster and H3K4me1 enrichment at the ATI region. Independent of ALKATI expression, H3K27ac enrichment (a histone mark characteristic of active promoters and enhancers47) was found in all analyzed melanoma samples, but not in the control lung cancer cell lines or in the 17 non-melanoma cell lines in ENCODE (FIGS. 11E and 11F). By integrating ChIP, DNase I hypersensitivity, and 5′-RACE data, the proximal cis-regulatory region was defined as chr2:29,445,000-29,447,100 and the potential transcription factor binding motifs were bioinformatically determined.48 (FIG. 11H). To test if the LTR could function as a promoter, a luciferase reporter assay was used, and it was found that, in contrast to lung cancer cell lines, melanoma cell lines showed low but consistent luciferase activity (FIG. 11G). Taken together, these data suggest that the H3K27ac mark at the ATI site might prime melanomas for ALKATI expression, which is consistent with the higher frequency of ALKATI in melanomas compared to other cancer types. However, these data also indicate that full activation of ALKATI expression requires additional trans-activating factors.
  • 5′-RACE (5′-rapid amplification of cDNA ends) coupled with next-generation sequencing was used to determine the ALKATI transcript. The ATI site was mapped to a 25 base pairs (bps) region in intron 19, approximately 400 bp upstream of ALK exon 20 (FIGS. 1B and 6A-C). The novel ALK transcript has a size of approximately 2500 bps (FIG. 7A). The ALKATI transcript has three predicted in-frame start codons (ATGs) resulting in proteins with molecular weights (MW) of 61.1 kDa (552 amino acids), 60.8 kDa (550 amino acids) and 58.7 kDa (532 amino acids) (FIGS. 6D and 6E). All three proteins maintain the intracellular tyrosine kinase domain, but lack the extracellular and transmembrane domains of ALKWT(FIG. 2A).
  • Immunoblots of two ALKWT-expressing neuroblastoma cell lines and two EML4-ALK variant-expressing lung cancer cell lines showed proteins at the expected MW of ˜220 kDa for ALKWT (and a smaller cleavage product lacking part of the extracellular region49) and of ˜120 kDa and ˜90 kDa for two EML4-ALK variants. ALKATI-expressing tumors revealed a double band at ˜60 kDa, suggesting that ALKATI is translated from more than one start codon (FIG. 2B). To experimentally confirm the predicted start codons, the three start codons were mutated individually or in combination and expressed them in 293T cells. Immunoblots revealed that each of the mutant ALKATI lost the corresponding protein band, indicating that all three start codons in ALKATI are functional and give rise to three distinct proteins (FIG. 2C).
  • The ALKATI proteins were phosphorylated in tumors with endogenous ALKATI expression and in cells with exogenous ALKATI expression (FIGS. 2B and 2C) indicating that ALKATI is active. This was confirmed by an in vitro kinase assay (FIG. 12A). A kinase-dead-ALKATI(ALKATI-KD), in which a lysine in the ATP-binding site of the kinase domain was replaced by a methionine15, was not phosphorylated or active. Reasoning that ALKATI may auto-activate by forming homodimers as do other RTKs50, the ability of self-interaction was tested using co-immunoprecipitation with V5- or HAtagged ALKATI proteins. The V5-ALKATI readily co-immunoprecipitated with the HA-ALKATI and vice versa, indicating that ALKATI self-interacts resulting in autophosphorylation and kinase activation (FIG. 2D). Using immunofluorescence, ALKATI was detected in both the nucleus and the cytoplasm, whereas ALKF1174L and EML4-ALK were found in the cytoplasm or the cell membrane (FIG. 2E). ALK immunohistochemistry in clinical samples confirmed the nuclear and cytoplasmic localization of ALKATI, suggesting that nuclear ALK staining in immunohistochemistry could serve as a clinical biomarker to identify patients with ALKATI-expressing tumors (FIGS. 2F and 12B).
  • Based on the analysis of the GTEx RNA-seq dataset, ALKATI is not expressed in normal tissues. To establish the functional consequences of ALKATI expression, Ba/F3, NIH-3T3, and melan-a cells were stably transduced with ALKATI, negative controls (ALKATI, empty vector), and positive controls (the oncogenic ALK variants ALKF1174L, EML4-ALK, and ALKWT, which was previously shown to be sufficient to drive oncogenesis at high endogenous expression levels6,12,58-61). In Ba/F3 cells, ALKATI expression led to IL-3-independent cell growth, as did the positive controls, but not the negative controls (FIG. 3A). In ALK-transformed Ba/F3 cells growing in media without IL-3, it was confirmed that ALKATI was expressed at a similar level compared to human tumors and that all ALK isoforms were phosphorylated and therefore active (FIG. 3B). The ALK dependency of IL-3-independent growth was reflected in the selection of Ba/F3 cells expressing greenfluorescent-protein (GFP), which was co-expressed from the ALK expression vectors (FIG. 12C). Consistent with the in vitro data, ALKATI-expressing NIH-3T3 and melan-a cells efficiently induced tumor growth in SCID mice (FIGS. 3C and 12D-12F).
  • In summary, all of the ALK-variant expressing cells (ALKATI, ALKF1174L, EML4-ALK, ALKWT) were able to establish growth factor-independent proliferation and tumorigenesis, with similar growth rates once the tumors were established. Importantly, the observed oncogenic capacity of ALKWT is consistent with previous reports that high endogenous expression or amplification of ALKWT drives oncogenesis and confers sensitivity to ALK inhibitors in neuroblastomas10,12, 58-61. To further explore the pathogenic role of ALKATI expression levels, NIH-3T3 cells were stably transduced with either a low or high titre of ALKATI resulting in cells expressing ALKATI at either low or at high levels. It was found that a further increase in ALKATI expression levels did not accelerate tumor graft formation and tumor growth indicating that ALKATI can drive tumourigenesis once a threshold of expression is reached (FIGS. 12G-12I).
  • To examine the therapeutic responses to pharmacologic ALK inhibition, Ba/F3 cells stably expressing various ALK isoforms were treated with three different ALK inhibitors (crizotinib, ceritinib, TAE-684). All ALK inhibitors effectively inhibited IL-3-independent growth of ALK-transformed Ba/F3 cells, whereas they had no effect on growth in the presence of IL-3 (FIGS. 4A and 13A-13B). Crizotinib inhibited ALKATI phosphorylation and downstream signaling in a concentration-dependent manner, further corroborating that ALKATI is activated through auto-phosphorylation (FIGS. 4B and 13E-13E). Crizotinib treatment induced also regression of ALKATI driven NIH-3T3-tumors in vivo, and immunohistochemistry of explanted tumors confirmed reduced cell proliferation, increased apoptosis, and inhibition of several oncogenic signaling pathways (FIGS. 4C-4E and 14A-14F).
  • Based on this encouraging pre-clinical data, a patient with metastatic melanoma with ALKATI expression was identified (FIGS. 4F and 4G). The patient had previously progressed on the combination of ipilimumab and nivolumab immunotherapy in a clinical trial, followed by palliative radiation and then dacarbazine chemotherapy. The compassionate use of crizotinib resulted in marked symptomatic improvement and tumor shrinkage within 6 weeks of therapy (FIG. 4H). This patient case provides additional evidence that ALKATI confers gain-of-function in patients and is amenable to pharmacologic targeting, warranting further clinical investigation.
  • Taken together, a novel ALK transcript, ALKATI, which arises independently of genomic aberrations at the ALK locus through alternative transcriptional initiation, was identified. ALKATI encodes shortened ALK proteins that are capable of driving oncogenesis in vitro and in vivo. ALKATI-driven tumors are sensitive to ALK inhibitors, suggesting that patients harboring such tumors could potentially benefit from ALK inhibitor therapy, but may not be identified using current clinical genomic assays, particularly those based on DNA sequencing. Importantly, alternative transcriptional initiation was discovered as a novel mechanism for oncogene activation, in addition to well-established genetic mechanisms such as mutations, translocations or amplifications. Other oncogenes may be activated via similar mechanisms in other human malignancies and their identification may provide new insights into oncogenesis and opportunities for therapeutic intervention.
  • 7. REFERENCES
    • 1. Consortium, G. T. The Genotype-Tissue Expression (GTEx) project. Nat Genet 45, 580-5 (2013).
    • 2. Lawrence, M. S. et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 499, 214-8 (2013).
    • 3. Lawrence, M. S. et al. Discovery and saturation analysis of cancer genes across 21 tumor types. Nature 505, 495-501(2014).
    • 4. Comprehensive molecular characterization of human colon and rectal cancer. Nature 487, 330-7 (2012).
    • 5. Comprehensive genomic characterization of squamous cell lung cancers. Nature 489, 519-25 (2012).
    • 6. Vogelstein, B. et al. Cancer genome landscapes. Science 339, 1546-58 (2013).
    • 7. Imielinski, M. et al. Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing. Cell 150, 1107-20 (2012).
    • 8. Weinstein, J. N. et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet 45, 1113-20 (2013).
    • 9. Anders, S., Reyes, A. & Huber, W. Detecting differential usage of exons from RNA-seq data. Genome Res 22, 2008-17 (2012).
    • 10. Mosse, Y. P. et al. Identification of ALK as a major familial neuroblastoma predisposition gene. Nature 455, 930-5 (2008).
    • 11. Chen, Y. et al. Oncogenic mutations of ALK kinase in neuroblastoma. Nature 455, 971-4 (2008).
    • 12. Janoueix-Lerosey, I. et al. Somatic and germline activating mutations of the ALK kinase receptor in neuroblastoma. Nature 455, 967-70 (2008).
    • 13. George, R. E. et al. Activating mutations in ALK provide a therapeutic target in neuroblastoma. Nature 455, 975-8 (2008).
    • 14. Morris, S. W. et al. Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non-Hodgkin's lymphoma. Science 263, 1281-4 (1994).
    • 15. Soda, M. et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature 448, 561-6 (2007).
    • 16. Lipson, D. et al. Identification of new ALK and RET gene fusions from colorectal and lung cancer biopsies. Nat Med 18, 382-4 (2012).
    • 17. Kouzarides, T. Chromatin modifications and their function. Cell 128, 693-705 (2007).
    • 18. Ram, O. et al. Combinatorial patterning of chromatin regulators uncovered by genome-wide location analysis in human cells. Cell 147, 1628-39 (2011).
    • 19. Wang, Z. et al. Combinatorial patterns of histone acetylations and methylations in the human genome. Nat Genet 40, 897-903 (2008).
    • 20. Mitelman, F., Johansson, B. & Mertens, F. The impact of translocations and gene fusions on cancer causation. Nature reviews. Cancer 7, 233-45 (2007).
    • 21. Cazes, A. et al. Characterization of rearrangements involving the ALK gene reveals a novel truncated form associated with tumor aggressiveness in neuroblastoma. Cancer Res 73, 195-204 (2013).
    • 22. Okubo, J. et al. Aberrant activation of ALK kinase by a novel truncated form ALK protein in neuroblastoma. Oncogene 31, 4667-76 (2012).
    • 23. Horn, S. et al. TERT promoter mutations in familial and sporadic melanoma. Science 339, 959-61 (2013).
    • 24. Huang, F. W. et al. Highly recurrent TERT promoter mutations in human melanoma. Science 339, 957-9 (2013).
    • 25. Lappalainen, T. et al. Transcriptome and genome sequencing uncovers functional variation in humans. Nature 501, 506-11 (2013).
    • 26. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078-9 (2009).
    • 27. Trapnell, C., Pachter, L. & Salzberg, S. L. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25, 1105-11 (2009).
    • 28. Sboner, A. et al. FusionSeq: a modular framework for finding gene fusions by analyzing paired-end RNA-sequencing data. Genome Biol 11, R104 (2010).
    • 29. McKenna, A. et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res 20, 1297-303 (2010).
    • 30. Robinson, J. T. et al. Integrative genomics viewer. Nat Biotechnol 29, 24-6 (2011).
    • 31. Carbon, S. et al. AmiGO: online access to ontology and annotation data. Bioinformatics 25, 288-9 (2009).
    • 32. Chi, P. et al. ETV1 is a lineage survival factor that cooperates with KIT in gastrointestinal stromal tumors. Nature 467, 849-53 (2010).
    • 33. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol 9, R137 (2008).
    • 34. Won, H. H., Scott, S. N., Brannon, A. R., Shah, R. H. & Berger, M. F. Detecting somatic genetic alterations in tumor specimens by exon capture and massively parallel sequencing. J Vis Exp, e50710 (2013).
    • 35. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754-60 (2009).
    • 36. DePristo, M. A. et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet 43, 491-8 (2011).
    • 37. Cibulskis, K. et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol 31, 213-9 (2013).
    • 38. Rausch, T. et al. DELLY: structural variant discovery by integrated paired-end and split-read analysis. Bioinformatics 28, i333-i339 (2012).
    • 39. Geiss, G. K. et al. Direct multiplexed measurement of gene expression with color-coded probe pairs. Nat Biotechnol 26, 317-25 (2008).
    • 40. Bennett, D. C., Cooper, P. J. & Hart, I. R. A line of non-tumorigenic mouse melanocytes, syngeneic with the B16 melanoma and requiring a tumor promoter for growth. Int J Cancer 39, 414-8 (1987).
    • 41. Refaeli, Y., Van Parijs, L., Alexander, S. I. & Abbas, A. K. Interferon gamma is required for activation-induced death of T lymphocytes. J Exp Med 196, 999-1005 (2002).
    • 42. Ponomarev, V. et al. A novel triple-modality reporter gene for whole-body fluorescent, bioluminescent, and nuclear noninvasive imaging. Eur J Nucl Med Mol Imaging 31, 740-51 (2004).
    • 43. Northcott, P. A. et al. Enhancer hijacking activates GFI1 family oncogenes in medulloblastoma. Nature 511, 428-434 (2014).
    • 44. Karolchik, D. et al. The UCSC Table Browser data retrieval tool. Nucleic Acids Res 32, D493-496 (2004).
    • 45. Xie, M. et al. DNA hypomethylation within specific transposable element families associates with tissue-specific enhancer landscape. Nat Genet 45, 836-841 (2013).
    • 46. Consortium, E. P. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57-74 (2012).
    • 47. Calo, E. & Wysocka, J. Modification of enhancer chromatin: what, how, and why? Mol Cell 49, 825-837 (2013).
    • 48. Grant, C. E., Bailey, T. L. & Noble, W. S. FIMO: scanning for occurrences of a given motif. Bioinformatics 27, 1017-1018 (2011).
    • 49. Moog-Lutz, C. et al. Activation and inhibition of anaplastic lymphoma kinase receptor tyrosine kinase by monoclonal antibodies and absence of agonist activity of pleiotrophin. J Biol Chem 280, 26039-26048 (2005).
    • 50. Lemmon, M. A. & Schlessinger, J. Cell signaling by receptor tyrosine kinases. Cell 141, 1117-1134 (2010).
    • 51. Reis, P. P. et al. mRNA transcript quantification in archival samples using multiplexed, color-coded probes. BMC Biotechnol 11, 46 (2011).
    • 52. Mathelier, A. et al. JASPAR 2014: an extensively expanded and updated open-access database of transcription factor binding profiles. Nucleic Acids Res 42, D142-147 (2014).
    • 53. Krueger, F. & Andrews, S. R. Bismark: a flexible aligner and methylation caller for Bisulfite-Seq applications. Bioinformatics 27, 1571-1572 (2011).
    • 54. Boeva, V. et al. Control-free calling of copy number alterations in deep-sequencing data using GC-content normalization. Bioinformatics 27, 268-269 (2011).
    • 55. Wang, J. et al. CREST maps somatic structural variation in cancer genomes with base-pair resolution. Nat Methods 8, 652-654 (2011).
    • 56. McLaren, W. et al. Deriving the consequences of genomic variants with the Ensembl API and SNP Effect Predictor. Bioinformatics 26, 2069-2070 (2010).
    • 57. Krzywinski, M. et al. Circos: an information aesthetic for comparative genomics. Genome Res 19, 1639-1645 (2009).
    • 58. Schulte, J. H. et al. High ALK receptor tyrosine kinase expression supersedes ALK mutation as a determining factor of an unfavorable phenotype in primary neuroblastoma. Clin Cancer Res 17, 5082-5092 (2011).
    • 59. Passoni, L. et al. Mutation-independent anaplastic lymphoma kinase overexpression in poor prognosis neuroblastoma patients. Cancer Res 69, 7338-7346 (2009).
    • 60. Bresler, S. C. et al. Differential inhibitor sensitivity of anaplastic lymphoma kinase variants found in neuroblastoma. Sci Transl Med 3, 108-114 (2011).
    • 61. Montavon, G. et al. Wild-type ALK and activating ALK-R1275Q and ALKF1174L mutations upregulate Myc and initiate tumor formation in murine neural crest progenitor cells. Oncotarget 5, 4452-4466 (2014).
    • 62. Wiesner, T. et al. Kinase fusions are frequent in Spitz tumors and spitzoid melanomas. Nat Commun 5, 3116 (2014)
  • Various publications are cited herein, the contents of which are hereby incorporated by reference in their entireties.

Claims (11)

1.-18. (canceled)
19. A method of determining whether an anti-cancer effect is likely to be produced in a cancer by an ALK inhibitor, comprising determining whether one or more cell of the cancer contains a detectable TALK, wherein the presence of a detectable TALK in the cell indicates that an ALK inhibitor would have an anti-cancer effect on the cancer.
20. A method of treating a subject having a cancer comprising: (a) determining whether a subject is likely to obtain therapeutic benefit from an ALK inhibitor, comprising determining whether one or more cancer cell of the subject contains a detectable TALK, wherein the presence of a detectable TALK in the cancer cell of the subject indicates that the subject is likely to benefit from an ALK inhibitor; and (b) treating the subject who is likely to benefit from an ALK inhibitor with a therapeutic amount of an ALK inhibitor.
21. The method of claim 20, comprising treating the subject with an alternative therapy other than an ALK inhibitor if no detectable TALK is present in the cancer cell of the subject.
22. The method of claim 20, wherein the cancer is selected from the group consisting of melanoma, thyroid carcinoma, lung adenocarcinoma, lung squamous cell carcinoma, renal clear cell carcinoma, and breast cancer.
23. The method of claim 20, wherein the ALK inhibitor is selected from the group consisting of crizotinib, ceritinib, NVP-TAE684, alectinib; AP26113, ASP-3026, CEP-37440, NMS-E628, PF-06463922, TSR-011, RXDX-101, and X-396.
24. A kit for determining whether an anti-cancer effect is likely to be produced in a cancer by an ALK inhibitor, comprising a means for determining the presence of a detectable TALK in one or more cell of the cancer.
25. The kit of claim 24, comprising means for determining the level of a TALK mRNA transcript, a TALK cDNA molecule corresponding thereto, and/or a TALK polypeptide encoded thereby in one or more cell of the cancer.
26. The kit of claim 25, wherein the means for determining the level of a TALK mRNA transcript is selected from the group consisting of probe hybridization, polymerase chain reaction (PCR), Northern blot, sequencing, microarray, and combinations thereof.
27. The kit of claim 25, wherein the means for determining the level of a TALK cDNA molecule is PCR.
28. The kit of claim 25, wherein the means for determining the level of a TALK polypeptide is selected from the group consisting of antibody binding, immunohistochemistry, Western blot, a functional assay, enzyme linked immunosorbent assay (ELISA), radioimmunoassays (RIA), enzyme immunoassays (EIA), mass spectrometry, 1-D or 2-D gel-based analysis system, immunoprecipitation, and combinations thereof.
US16/258,477 2014-04-10 2019-01-25 Novel isoform of anaplastic lymphoma kinase and its uses Abandoned US20190292529A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/258,477 US20190292529A1 (en) 2014-04-10 2019-01-25 Novel isoform of anaplastic lymphoma kinase and its uses

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201461978106P 2014-04-10 2014-04-10
PCT/US2015/025289 WO2015157624A2 (en) 2014-04-10 2015-04-10 A novel isoform of anaplastic lymphoma kinase and its uses
US15/288,531 US10190104B2 (en) 2014-04-10 2016-10-07 Isoform of anaplastic lymphoma kinase and its uses
US16/258,477 US20190292529A1 (en) 2014-04-10 2019-01-25 Novel isoform of anaplastic lymphoma kinase and its uses

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/288,531 Division US10190104B2 (en) 2014-04-10 2016-10-07 Isoform of anaplastic lymphoma kinase and its uses

Publications (1)

Publication Number Publication Date
US20190292529A1 true US20190292529A1 (en) 2019-09-26

Family

ID=54288551

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/288,531 Expired - Fee Related US10190104B2 (en) 2014-04-10 2016-10-07 Isoform of anaplastic lymphoma kinase and its uses
US16/258,477 Abandoned US20190292529A1 (en) 2014-04-10 2019-01-25 Novel isoform of anaplastic lymphoma kinase and its uses

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US15/288,531 Expired - Fee Related US10190104B2 (en) 2014-04-10 2016-10-07 Isoform of anaplastic lymphoma kinase and its uses

Country Status (6)

Country Link
US (2) US10190104B2 (en)
EP (2) EP3129477B1 (en)
AU (1) AU2015243241A1 (en)
CA (1) CA2945324A1 (en)
ES (1) ES2811927T3 (en)
WO (1) WO2015157624A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107022602A (en) * 2016-02-02 2017-08-08 生物梅里埃公司 A kind of method of detection ALK genetic recombination

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170356918A1 (en) * 2014-12-03 2017-12-14 Ignyta, Inc. Multiplexed immunohistochemistry assays for diagnosis and treatment of cancer

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007059300A2 (en) * 2005-11-15 2007-05-24 Medimmune, Inc. Anti-alk antagonist and agonist antibodies and uses thereof
US8168383B2 (en) * 2006-04-14 2012-05-01 Cell Signaling Technology, Inc. Gene defects and mutant ALK kinase in human solid tumors
EP2042191B1 (en) * 2007-09-28 2013-01-02 Roberto Chiarle Anaplastic Lymphoma Kinase (ALK) as oncoantigen for lymphoma vaccination
US8383793B2 (en) * 2010-04-15 2013-02-26 St. Jude Children's Research Hospital Methods and compositions for the diagnosis and treatment of cancer resistant to anaplastic lymphoma kinase (ALK) kinase inhibitors
WO2012019132A2 (en) * 2010-08-06 2012-02-09 Cell Signaling Technology, Inc. Anaplastic lymphoma kinase in kidney cancer
KR101264810B1 (en) * 2010-11-25 2013-05-15 한국과학기술연구원 Method for Screening of ALK gene rearrangement and diagnosis of cancer using the same
WO2013059740A1 (en) * 2011-10-21 2013-04-25 Foundation Medicine, Inc. Novel alk and ntrk1 fusion molecules and uses thereof

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107022602A (en) * 2016-02-02 2017-08-08 生物梅里埃公司 A kind of method of detection ALK genetic recombination

Also Published As

Publication number Publication date
WO2015157624A3 (en) 2015-12-10
WO2015157624A2 (en) 2015-10-15
EP3735973A1 (en) 2020-11-11
EP3129477A4 (en) 2017-11-01
US10190104B2 (en) 2019-01-29
CA2945324A1 (en) 2015-10-15
AU2015243241A1 (en) 2016-11-03
US20170022485A1 (en) 2017-01-26
EP3129477B1 (en) 2020-05-20
EP3129477A2 (en) 2017-02-15
ES2811927T3 (en) 2021-03-15

Similar Documents

Publication Publication Date Title
Wiesner et al. Alternative transcription initiation leads to expression of a novel ALK isoform in cancer
Indraccolo et al. Genetic, epigenetic, and immunologic profiling of MMR-deficient relapsed glioblastoma
Pemov et al. Low mutation burden and frequent loss of CDKN2A/B and SMARCA2, but not PRC2, define premalignant neurofibromatosis type 1–associated atypical neurofibromas
Saarenheimo et al. The value of liquid biopsies for guiding therapy decisions in non-small cell lung cancer
Nanba et al. Molecular heterogeneity in aldosterone-producing adenomas
Paik et al. Next-generation sequencing of stage IV squamous cell lung cancers reveals an association of PI3K aberrations and evidence of clonal heterogeneity in patients with brain metastases
Young et al. Analysis of VHL gene alterations and their relationship to clinical parameters in sporadic conventional renal cell carcinoma
Vanni et al. The current state of molecular testing in the BRAF-mutated melanoma landscape
Wu et al. Identification of targetable FGFR gene fusions in diverse cancers
Pfister et al. BRAF gene duplication constitutes a mechanism of MAPK pathway activation in low-grade astrocytomas
Felsberg et al. Prognostic significance of molecular markers and extent of resection in primary glioblastoma patients
JP2020100641A (en) Methods of treating cancer
Garcia-Carracedo et al. Loss of PTEN expression is associated with poor prognosis in patients with intraductal papillary mucinous neoplasms of the pancreas
US20170198353A1 (en) Kras mutations and resistance to anti-egfr treatment
US20180327863A1 (en) Arid1a and ppp2r1a mutations in cancer
Wang et al. The molecular landscape of synchronous colorectal cancer reveals genetic heterogeneity
CA2862580A1 (en) Biomarkers of response to nae inhibitors
Oulhen et al. Circulating tumor cell copy-number heterogeneity in ALK-rearranged non-small-cell lung cancer resistant to ALK inhibitors
Biswas et al. Genomic profiling of multiple sequentially acquired tumor metastatic sites from an “exceptional responder” lung adenocarcinoma patient reveals extensive genomic heterogeneity and novel somatic variants driving treatment response
US20190292529A1 (en) Novel isoform of anaplastic lymphoma kinase and its uses
M. Naeini et al. Multi-omic features of oesophageal adenocarcinoma in patients treated with preoperative neoadjuvant therapy
US20210363588A1 (en) Addressing Treatments for Glioblastoma
Smit et al. High‐resolution ERG‐expression profiling on G eneChip exon 1.0 ST arrays in primary and castration‐resistant prostate cancer
CA3198134A1 (en) Immunotherapy response signature
US11697845B2 (en) Directing treatments for glioblastoma based on identifying a somatic structural rearrangement upstream from TERT gene

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO PAY ISSUE FEE