AU2013295805A1 - Fusion proteins and methods thereof - Google Patents

Fusion proteins and methods thereof

Info

Publication number
AU2013295805A1
AU2013295805A1 AU2013295805A AU2013295805A AU2013295805A1 AU 2013295805 A1 AU2013295805 A1 AU 2013295805A1 AU 2013295805 A AU2013295805 A AU 2013295805A AU 2013295805 A AU2013295805 A AU 2013295805A AU 2013295805 A1 AU2013295805 A1 AU 2013295805A1
Authority
AU
Australia
Prior art keywords
fgfr
fgfr3
fusion
tacc3
tacc
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.)
Granted
Application number
AU2013295805A
Other versions
AU2013295805B2 (en
Inventor
Antonio Iavarone
Anna Lasorella
Raul Rabadan
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.)
Columbia University in the City of New York
Original Assignee
Columbia University in the City of New York
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 Columbia University in the City of New York filed Critical Columbia University in the City of New York
Priority claimed from PCT/US2013/051888 external-priority patent/WO2014018673A2/en
Publication of AU2013295805A1 publication Critical patent/AU2013295805A1/en
Application granted granted Critical
Publication of AU2013295805B2 publication Critical patent/AU2013295805B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Description

FUSION PROTEINS AND METHODS THEREOF 000 j This application claims the benefit of and priority to U.S. Provisional Patent Application No. 61/675,006, filed on July 24, 2012, the content of which is hereby incorporated by reference in its entirety.
[0(502] All patents, paieni applications and publications cited herein are hereby incorporated by reference in their entirety. The disclosures of these publications in their entireties are hereby incorporated by reference into this application.
[0003] This patent disclosure contains material that is subject to copyright protection. The copyright owner has no objection to the facsimile reproduction by anyone of the patent document or the paieni disclosure as it appears in the U.S. Paieni and Trademark Office patent file or records, but otherwise reserves any and all copyright rights.
GOVERNMENT SUPPORT
[0(584] This invention was made with government support under Grant Nos.
R01CA101644 and R01CA085628 awarded by the Naiional Cancer Institute. The
Government has certain rights in the invention.
BACKGROUND OF THE INVENTION
[0005] Glioblas toma multiforme (GBM) is the most common form of brain cancer and among the most incurable and lethal of all human cancers. The current standard of care includes surgery, chemotherapy, and radiation therapy. However, the prognosis of GBM remains uniformly poor. There are few available targeted therapies and none that specifically target GBM.
[0006] The target population of GBM patients who may cany FGFR-TACC gene fusions and would benefit from targeted inhibition of FGFR kinase activity is estimated to correspond to 6,000 patients per year world-wide.
SUMMARY OF THE INVENTION
[0(5(17] The invention is based, at least in part, on the disco very of a highly expressed class of gene fissions in GBM, which join the tyrosine kinase domain of FGFR genes to the TACC domain of TACC1 or TACC3. The invention is based, at least in part, on the finding
- I - that FGFR-TACC fusions identify a subset of GBM patients who will benefit from targeted inhibition of the tyrosine kinase activity of FGFR. Identification of fusions of FGFR and TACC genes in glioblastoma patients and other subjects afflicted with a gene-fusion associated cancer (such as an epithelial cancer) are useful therapeutic targets.
[ΘΘ08] An aspect of the invention provides for a purified fusion protein comprising a tyrosine kinase domain of an FGFR protein fused to a polypeptide that constitutively aciivates the tyrosine kinase domain of the FGFR protein. In one embodiment, the FGFR protein is FGFR1 , FGFR2, FGFR3, or FGR4. in another embodiment, the purified fusion protein is essentially free of other human proteins.
[©009] An aspect of the invention provides for a purified fusion protein comprising a transforming acidic coiled-coil (TACC) domain fused to a polypeptide with a tyrosine kinase domain, wherein the TACC domain constitutively activates the tyrosine kinase domain. In one embodiment, the TACC protein is TA.CC 1 , TACC2, or TACC3. In another embodiment, the purified fusion protein is essentially free of other human proteins.
[0010] An aspect of the invention provides for a purified fusion protein comprising the tyrosine kinase domain of an FGFR protein fused 5' to the TACC domain of a transforming acidic coiled-coil-containing (TACC) protein. In one embodiment, the FGFR protein is FGFRi, FGFR2, FGFR3, or FGR4. In another embodiment, the TACC protein is TACCL TACC2, or TACC3. In another embodiment, the purified fusion protein is essentially free of other hitman proteins.
[00 1] An aspect of the invention provides for a purified fusion protein encoded by an FGFRl-TACC l nucleic acid, wherein FGFR l-TACC l comprises a combination of exons 1 - 17 of FGFR I located on human chromosome 8 l 1 spliced 5' to a combination of exons 7-13 of TACCl located on human chromosome 8pl 1 , wherein a genomic breakpoint occurs in any one of exons 1-17 of FGFRI and any one of exons 7-13 of TACC L In another embodiment, the purified fusion protein is essentially free of other human proteins.
[0012] An aspect of the invention provides for a purified fusion protein encoded by an FGFR2-TACC2 nucleic acid, wherein FGFR2 -TACC2 comprises a combination of any exons 1-1 8 of FGFR2 located on human chromosome 1 Qq26 spliced 5' to a combination of any exons 1-23 of TACC2 located on human chromosome K)q26. In another embodiment, the purified fusion protein is essentially free of other human proteins. [0(513] An aspect of the invention provides for a purified fusion protein encoded by an FGFR3-TACC3 nucleic acid, wherein FGFR3-TACC3 comprises a combination of exons 1 - 16 of FOFR3 located on human chromosome 4 l6 spliced 5 ' to a combination of exons 8-16 of TACC3 located on human chromosome 4 l 6, wherein a genomic breakpoint occurs in any one of exons 1-16 of FGFR3 and any one of exons 8-16 of TACC3. In another embodiment, the purified fusion protein is essentially free of other human proteins.
[0(514] An aspect of (he invention provides for a purified fusion protein encoded by an FGFR3-TACC3 nucleic acid, wherein FGFR3-TACC3 comprises a combination of exons 1- 16 of FGFR3 located on human chromosome 4 l6 spliced 5 ' to a combination of exons 8-16 of TACC-3 located on human chromosome 4 l 6, wherein a genomic breakpoint occurs in any one οϊ introns 1-16 of FGFR3 and any one of exons 8- 16 of TAC-C3. In another embodiment, the purified fusion protein is essentially free of other human proteins.
[0(515] An aspect of the invention provides for a purified fusion protein encoded by an FGFR3-TACC3 nucleic acid, wherein FGFR3-TACC3 comprises a combination of exons 1- 16 of FGFR3 located on human chromosome 4 l6 spliced 5' to a combination of exons 8-16 of TACC3 located on human chromosome 4pl6, wherein a genomic breakpoint occurs in any one of exons 1-16 of FGFR3 and any one of introns 7-16 of TACC3. In another embodiment, the purified fusion protein is essentially free of other human proteins.
[0016] An aspect of the invention provides for a purified fusion protein encoded by an FGFR3-TACC3 nucleic acid, wherein FGFR3-TACC3 comprises a combination of exons 1- 16 of FGFR3 located on human chromosome 4 I6 spliced 5' to a combination of exons 8- 16 of TACC3 located on human chromosome 4pl6, wherein a genomic breakpoint occurs in any one of introns 1-16 of FGFR3 and any one of introns 7-16 of TACC-3. In another embodiment, the purified fusion protein is essentially free of other human proteins.
[0017] An aspect of the invention provides for a synthetic nucleic acid encoding the fusion proteins described above.
[0(518] An aspect of the invention provides for a purified FGFR3-TACC3 fusion protein comprising SEQ ID NO: 79, 158, 159, 160, or 161 , In another embodiment, the purified fusion protein is essentially free of other human proteins.
[0019] An aspect of the invention provides for a purified FGFR3-TACC3 fusion protein having a genomic breakpoint comprising at least 3 consecutive amino acids from amino acids 730-758 of SEQ ID NO: 90 and comprising at leasi 3 consecutive amino acids from amino acids 549-838 of SEQ ID NO: 92. in another embodiment, the purified fusion protein is essentially free of other human proteins.
[ΘΘ2Θ] An aspect of the invention provides for a purified FGFR3-TACC3 fusion protem having a genomic breakpoint comprising SEQ ID NO: 78. In another embodiment, the purified fusion protein is essentially free of other human proteins,
[0021] An aspect of the invention provides for a purified FGFR3-TACC3 fusion protein having a genomic breakpoint comprising any one of SEQ ID NOS: 85, 86, 87, or 89. In another embodiment, the purified fusion protein is essentially free of other human proteins.
[0022] An aspect of the invention provides for a purified FGFR1 -TACC1 fusion protein comprising SEQ ID NO: 150. In another embodiment, the purified fusion protein is essentially free of other human proteins.
[ΘΘ23] An aspect of the invention provides for a purified FGFRl-TACCl fusion protem having a genomic breakpoint comprising at least 3 consecutive amino acids from amino acids 746-762. of SEQ ID NO: 146 and comprising at least 3 consecutive amino acids from amino acids 572-590 of SEQ ID NO: 148. In another embodiment, the purified fusion protem is essentially free of other human proteins.
[8024] An aspect of the invention provides for a purified FGFRl-TACCl fusion protem having a genomic breakpoint comprising SEQ ID NO: 88, In another embodiment, the purified fusion protein is essentially free of other human proteins.
[0025] An aspect of the invention provides for a purified DNA encoding an FGFR3- TACC3 fusion protein comprising SEQ ID NO: 94. In another embodiment the purified fusion protein is essentially free of other human proteins.
[0026] An aspect of the invention provides for a synthetic nucleic acid encoding an FGFR3-TACC3 fusion protein having a genomic breakpoint comprising at least 9 consecutive in-frame nucleotides from nucleotides 2443-2530 of SEQ ID NO: 91 and comprising at least 9 consecutive in-frame nucleotides from nucleotides 1 800-2847 of SEQ ID NO: 93. [0027] An aspect of the invention provides for a synthetic nucleic acid encoding an FGFR3-TACC3 fusion protein having a genomic breakpoint comprising any one of SEQ ID NOS: 1 -77,
[0028] An aspect of the invention provides for a synthetic nucleic acid encoding an FGFRI -TACC I fusion protein comprising SEQ ID NO: 1 51.
{0029'J An aspect of the invention provides for a synthetic nucleic acid encoding an FGFRl -TACCl fusion protein having a genomic breakpoint comprising at least 9 consecutive in- frame nucleotides from nucleotides 3178-3228 of SEQ ID NO: 147 and comprising at least 9 consecutive in-frame nucleotides from nucleotides 2092-2794 of SEQ ID NO: 149.
[0030] An aspect of the invention provides for a synthetic nucleic acid encoding an FGFRl -TACC l fusion protein having a genomic breakpoint comprising SEQ ID NO: 83.
[0031] An aspect of the invention provides for an antibody or antigen-binding fragment thereof, that specifically binds to a purified fusion protein comprising a tyrosine kinase domain of an FGFR protein fused to a polypeptide that constitutively activates the tyrosine kinase domain of the FGFR protein. In one embodiment, the FGFR protein is FGFR1 , FGFR2, FGFR3, or FGFR4. In another embodiment, the fusion protein is an FGFR-TACC fusion protein. In a further embodiment, the FGFR-TACC fusion protein is FGFRl-TACC l , FGFR2-TACC2, or FGFR3 -TACC3. In some embodiments, the FGFRl -TACC l fusion protein comprises the amino acid sequence of SEQ ID NO: 350. In other embodiments, the FGFR3-TACC3 fusion protein comprises the amino acid sequence of SEQ ID NO: 79, 158, 159, 160, or 161 .
[0032] An aspect of the invention provides for a composition for decreasing in a subject the expression le vel or activity of a fusion protein comprising the tyrosine kinase domain of an FGFR protein fused to a polypeptide that constitutively activates the tyrosine kinase domain of the FGFR protein, the composition in an admixture of a pharmaceutically acceptable carrier comprising an inhibitor of the fission protein. In one embodiment, the fusion protein is an FGFR-TACC fusion protein. In another embodiment, the inhibitor comprises an antibody that specifically binds to a FGFR-TACC fusion protein or a fragment thereof; a small molecule that specifically binds to a FGFR protein; a small molecule that specifically binds to a TACC protein; an antisense RNA or antisense DNA that decreases expression of a FGFR-TACC fusion polypeptide; a siRNA that specifically targets a FGFR- TACC fusion gene; or a combination of the listed inhibitors. In a further embodiment, the FGFR protein is FGFRL FGFR2, FGFR.3, or FGFR4. In some embodiments, the FGFR- TACC fusion protein is FGFR l -TACC1 , F GFR2 -TACC2, or FGFR3-TACC3. In other embodidments, the small molecule that specifically binds to a FGFR protein comprises AZD4547, VP-BGJ398, PD173074, NF449, TK1258, BIBF-1 120, BMS-582664, AZD- 2171 , TSU68, AB 1010, AP24534, E-7080, LY2874455, or a combination of the listed small molecules.
[0033] An aspect of the invention provides for a method for decreasing in a subject in need thereof the expression level or activity of a fusion protein comprising the tyrosine kinase domain of an FGFR protein fused to a polypeptide that eonstitutively activates the tyrosine kinase domain of the FGFR protein. In one embodiment, the method comprises
administering to the subject a therapeutic amount of a composition for decreasing the expression level or activity in a subject of a fusion protein comprising the tyrosine kinase domain of an FGFR protein fused to a polypeptide that eonstitutively activates the tyrosine kinase domain of the FGFR protein. In one embodiment, the method comprises obtaining a sample from the subject to determine the level of expression of an FGFR fusion molecule in the subject. In some embodiments, the sample is incubated with an agent that binds to an FGFR fusion molecule, such as an antibody, a probe, a nucleic acid primer, and the like. In one embodiment, the detection or determining comprises nucleic acid sequencing, selective hybridization, selective amplification, gene expression analysis, or a combination thereof. In another embodiment, the detection or determination comprises protein expression analysis, for example by western blot analysis, ELISA, or other antibody detection methods. In a further embodiment, the method comprises determining whether the fusion protein expression level or activity is decreased compared to fusion protein expression level or activity prior to administration of the composition, thereby decreasing the expression level or activity of the fusion protein. In one embodiment, the fusion protein is an FGFR-TACC fusion protein. In a further embodiment, the FGFR protein is FGFRl, FGFR2, FGFR3, or FGFR4. In some embodiments, the FGFR-TACC fusion protein is FGFRl -TACC 1, FGFR2- TACC2, or FGFR3-TACC3. in one embodiment, the composition for decreasing the expression level or activity of a fusion protein comprises an antibody that specifically binds to a FGFR-TACC fusion protein or a fragment thereof; a small molecule that specifically binds to a FGFR protein; a small molecule that specifically binds to a TACC protein: an antisense RNA or antisense D A that decreases expression of a FGFR-TACC fusion polypeptide; a siRNA that specifically targets a FGFR-TACC fusion gene; or a combination of the listed inhibitors, in a further embodiment, the FGFR protein is FGFR] , FGFR2, FGFR3, or FGFR4. In some embodiments, the FGFR-TACC fusion protein is FGFR1- TACC1, FGFR2-TACC2, or FGFR3-TACC3. In other embodiments, the small molecule that specifically binds to a FGFR protein comprises AZD4547, NVP--BGJ398, PD 173074, NF449, TK1258, BIBF-1120, BMS-582664, AZD-2171 , TSU68, ABIOIO, AP24534, E-7080, LY2874455, or a combination of the small molecules listed.
[0034] An aspect of the invention provides for a method for treating a gene-fusion associated cancer in a subject in need thereof, the method comprising administering to the subject an effective amount of a FGFR fusion molecule inhibitor. In one embodiment, the gene-fusion associated cancer comprises an epithelial cancer. In one embodiment, the gene-fusion associated cancer comprises glioblastoma multiforme, breast cancer, lung cancer, prostate cancer, or colorectal carcinoma. In one embodiment, the method comprises obtaining a sample from the subject to determine the level of expression of an FGFR fusion molecule in the subject. Tn some embodiments, the sample is incubated with an agent that binds to an FGFR fusion molecule, such as an antibody, a probe, a nucleic acid primer, and the like. In one embodiment, the detection or determining comprises nucleic acid sequencing, selective hybridization, selective amplification, gene expression analysis, or a combination thereof. In another embodiment, the detection or determination comprises protein expression analysis, for example by western blot analysis, EL1SA, or other antibody detection methods. In another embodiment, the FGFR fusion protein comprises an FGFR protein fused to a polypeptide that constitufively activates the tyrosine kinase domain of the FGFR protein. In one embodiment, the fusion protein is an FGFR-TACC fusion protein. In another embodiment, the inhibitor comprises an antibody that specifically binds to a FGFR-TACC fusion protein or a fragment thereof; a small molecule that specifically binds to a FGFR protein a small molecule that specifically binds to a TACC protein; an antisense RNA or antisense DNA that decreases expression of a FGFR-TACC fusion polypeptide; a siRNA that specifically targets a FGFR-TACC fusion gene; or a combination of the listed inhibitors, in a further embodiment, the FGFR protein is FGFRi, FGFR2, FGFR3, or FGFR4. In some embodiments, the FGFR-TACC fusion protein is FGFRi-TACCi, FGFR2-TACC2, or FGFR3-TACC3. In other embodiments, the small molecule that specifically binds to a FGFR protein comprises AZD4547, NVP-BGJ398, PD 173074, NF449, TK1258, BTBF- 1 ί 20, BMS- 582664, AZD-2171, TSU68, ABI010, AP24534, E-7080, LY2874455, or a combination of the small molecules listed.
[003S] An aspect of the invention provides for a method of decreasing growth of a solid tumor in a subject in need thereof, the method comprising administering to the subject an effective amount of a FGFR fusion molecule inhibitor, wherein the inhibitor decreases the size of the solid tumor, in one embodiment, the sol id tumor comprises gl ioblastoma multiforme, breast cancer, lung cancer, prostate cancer, or colorectal carcinoma. In one embodiment, the method comprises obtaining a sample from the subject to determine the level of expression of an FGFR fusion molecule in the subject. In some embodiments, the sample is incubated with an agent that binds to an FGFR fusion molecule, such as an antibody, a probe, a nucleic acid primer, and the like. In one embodiment, the detection or determining comprises nucleic acid sequencing, selective hybridization, selective amplification, gene expression analysis, or a combination thereof. In another embodiment, the detection or determination comprises protein expression analysis, for example by western blot analysis, ELISA, or other antibody detection methods. In another embodiment, the FGFR fusion protein comprises an FGFR protein fused to a polypeptide that constitutively activates the tyrosine kinase domain of the FGFR protein, in one embodiment, the fusion protein is an FGFR-TACC fusion protein. In another embodiment, the inhibitor comprises an antibody that specifically binds to a FGFR-TACC fusion protein or a fragment thereof; a small molecule that specifically binds to a FGFR protein; a small molecule that specifically binds to a TACC protein; an antisense R A or antisense DNA that decreases expression of a FGFR-TACC fusion polypeptide; a siRNA that specifically targets a FGFR-TACC fusion gene; or a combination of the listed inhibitors. In a further embodiment, the FGFR protein is FGFRI, FGFR2, FGFR3, or FGFR4. in some embodiments, the FGFR-TACC fusion protein is FGFR I-TACCI, FGFR2-TACC2, or FGFR3-TACC3. In other embodiments, the small molecule that specifically binds to a FGFR protein comprises AZD4547, NVP- BGJ398, PD173074, NF449, TKI258, BlBF-1 120, BMS-582664, AZD-2171, TSU68, ΑΒ10Ϊ 0, AP24534, E-7080, LY2874455, or a combination of the small molecules listed. Θί136| An aspect of the invention provides for a diagnostic kit for determining whether a sample from a subject exhibits a presence of a FGFR fusion, the kit comprising at least one oligonucleotide that specifically hybridizes to a FGFR fusion, or a portion thereof. In one embodiment, the oligonucleotides comprise a set of nucleic acid primers or in situ hybridization probes. In another embodiment, the oligonucleotide comprises SEQ ID NO: 162, 163, 164, 165, 166, 167, 168, 169, or a combination of the listed oligonucleotides. In one embodiment, the primers prime a polymerase reaction only when a FGFR fusion is present. In another embodiment, the determining comprises gene sequencing, selective hybridization, selective amplification, gene expression analysis, or a combination thereof. In a further embodiment, the FGFR-fusion is an FGFR-TACC fusion. In some embodiments, the FGFR is FGFRI, FGFR2, FGFR3, or FGFR4. In other embodiments, the FGFR-TACC fusion is FGFRI -TACC 1 , FGFR2-TACC2, or FGFR3 -TACC3.
[t>837] An aspect of the invention provides for a diagnostic kit for determining whether a sample from a subject exhibits a presence of a FGFR fusion protein, the kit comprising an antibody that specifically binds to a FGF fusion protein comprising SEQ ID NO: 79, 85, 86, 87, 88, 89, 150, 158, 159, 160, or 161, wherein the antibody will recognize the protein only when a FGFR fusion protein is present. In one embodiment, the antibody directed to and FGFR fusion comprising SEQ ID NO: 79, 85, 86, 87, 88, 89, 150, 158, 159, 160, or 161. In a further embodiment, the FGFR-fusion is an FGFR-TACC fusion. In some embodiments, the FGFR is FGFRI, FGFR2, FGFR3, or FGFR4. In other embodiments, the FGFR-TACC fusion is FGFR1-TACC1, FGFR2-TACC2, or F GFR3 -T.ACC 3.
[0(538] An aspect of the invention provides for a method for detecting the presence of a FGFR fusion in a human subject. In one embodiment, the method comprises obtaining a biological sample from the human subject. In some embodiments, the sample is incubated with an agent that binds to an FGFR fusion molecule, such as an antibody. In another embodiment, the detection or determination comprises protein expressio analysis, for example by western blot analysis, ELISA, or other antibody detection methods. In some embodiments, the method further comprises assessing whether to administer a FGFR fusion molecule inhibitor based on the expression pattern of the subject. In further embodiments, the method comprises administering a FGFR fusion molecule inhibitor to the subject. In another embodiment, the method comprises defecting whether or not there is a FGFR fusion present in the subject, in one embodiment, the detecting comprises measuring FGFR fusion protein levels by ERISA using an antibody directed to SEQ ID NO: 79, 85, 86, 87, 88, 89, 150, 158, 159, 160, or 161 ; western blot using an antibody directed to SEQ ID NO: 79, 85, 86, 87, 88, 89, 150, 158, 159, 160, or 161; mass spectroscopy, isoelectric focusing, or a combination of the listed methods. In some embodiments, the FGFR-fusion is an FGFR- TACC fusion. In other embodiments, the FGFR is FGFR1 , FGFR2, FGFR3, or FGFR4. In other embodiments, the FGFR- TACC fusion is FGFRl -TACC 1, FGFR2-TACC2, or FGFR3- TACC3.
[t>839] An aspect of the invention provides for a method for detecting the presence of a FGFR fusion in a human subject. In one embodiment, the method comprises obtaining a biological sample from a human subject. In some embodiments, the sample is incubated with an agent that binds to an FGFR fusion molecule, such as a probe, a nucleic acid primer, and the like. In other embodiments, the detection or determination comprises nucleic acid sequencing, selective hybridization, selective amplification, gene expression analysis, or a combination thereof. In some embodiments, the method further comprises assessing whether to administer a FGFR fusion molecule inhibitor based on the expression pattern of the subject. In further embodiments, the method comprises administering a FGFR fusion molecule inhibitor to the subject. In another embodiment, the method comprises detecting whether or not there is a nucleic acid sequence encoding a FGFR fusion protein in the subject, in one embodiment, the nucleic acid sequence comprises any one of SEQ ID NOS: 1-77, 80-84, or 95- 145. In another embodiment, the detecting comprises using hybridization, amplification, or sequencing techniques to detect a FGFR fusion. In a further embodiment, the amplification uses primers comprising SEQ ID NO: 162, 163, 164, 165, 166, 167, 168, or 169. In some embodiments, the FGFR-fusion is an FGFR-TACC fusion, in other embodiments, the FGFR is FGFR I, FGFR2, FGFR3, or FGFR4. In other embodiments, the FGFR-TACC fusion is FGFRl -TACC I, FGFR2-TACC2, or FGFR3-TACC3.
[Θ04Θ] An aspect of the invention provides for a method of initiating oncogenic transformation in vitro. The method comprises (a) transducing cells cultured in vitro with FGFR-TACC fusion DNA; and (b) determining whether the ceils acquire the ability to grow in anchorage-independent conditions, form multi-layered foci, or a combination thereof [0041] An aspect of the invention provides for a method of initiating oncogenic transformation in vivo. The method comprises (a) transducing ceils cultured in vitro with FGFR-TACC fusion DNA; (b) injecting a mouse with the transduced cells; and (c) determining whether a tumor grows in the mouse. Tn one embodiment, the injecting is a subcutaneous or intracranial injection.
[0(542] An aspect of the invention provides a method of identifying a compound that decreases the oncogenic activity of a FGFR-TACC fusion. The method comprises (a) transducing a cell cultured in vitro with FGFR-TACC DNA ; (b) contacting a cell with a ligand source for an effective period of time; and (c) determining whether the cells acquire the ability to grow in anchorage-independent conditions, form multi-layered foci, or a combination thereof, compared to cells c ultured in the absence of the test compound.
BRIEF DESCRIPTION OF THE FIGURES
[0043] To conform to the requirements for PCT patent applications, many of the figures presented herein are black and white representations of images originally created in color. In the below descriptions and the examples, the colored plots, and images are described in terms of its appearance in black and white. The original color versions can be viewed in Singh el al., Science (2012), 337(6099): 1231-5 (including the accompanying Supplementary
Information available in the on-line version of the manuscript available on the Science web site). For the purposes of the PCT, the contents of Singh et al., Science (2012),
337(6099): 1231-5, including the accompanying "Supplementary Information,'" are herein incorporated by reference.
[0044] FIG, A is a graph that shows genes recurrently involved in gene fusions in TCGA. Only genes involved in at least three gene fusions across different samples are displayed.
[0045] FIG. IB shows an FGFR3-TACC3 gene fusion identified by whole transcriptome sequencing of GSCs. 76 split-reads (SEQ ID NOS: 2-77, respectively) are shown aligning on the breakpoint. The predicted reading frame at the breakpoint is shown at the top (FGFR3 nucleotide sequence in purple (left) and TACC3 nucleotide sequence in green (right); SEQ ID NO: 1 ) with FGFR3 sequences below the predicted reading frame in red (left) and TACC3 in blue (right). The puiative amino acid sequence (SEQ ID NO: 78) corresponding to SEQ ID NO: I is shown above the predicted reading frame. [0(546] FIG. 1C shows an FGFR3-TACC3 gene fusion identified by whole transcriptome sequencing of GSCs. On the left, FGFR3-TACC3 -specific PGR from cD A derived from GSCs and GBM is shown. On the right, Sanger sequencing chromatograra shows the reading frame at the breakpoint (SEQ ID NO: 80) and putative translation of the fusion protein (SEQ ID NO: 85) in the positive samples.
{0047] FIG, ID shows an FGFR3-TACC3 gene fusion identified by whole transcriptome sequencing of G SCs. Amino acid sequence of the FGFR3-TACC3 protein is shown (SEQ ID NO: 79). Residues corresponding to FGFR3 or to TACC3 are shown in green or red (underlined), respectively. The fusion protein joins the tyrosine kinase domain of FGFR3 to the TACC domain of TACC3.
[0048] FIG. IE shows an FGFR3-TACC3 gene fusion identified by whole transcriptome sequencing of GSCs. Genomic fusion of FGFR3 exon 17 with intron 7 of TACC3 is shown. In the fused mRNA, exon 16 of FGFR3 is spliced 5' to exon 8 of TACC3. Filled arrows indicate the position of the fusion-genome primers, which generate fusion- specific PGR products in GSC-1 123 and GBM- 1 123.
[0049] FIG. 2A shows recurrent gene fissions bet een FGFR and TACC genes in GBM. Specifically, FGFR3-TACC3 gene fusions are shown that were identified by exome sequencing analysis. Split-reads are shown aligning the genomic breakpoints of FGFR3 and TACC3 genes in the four TCGA GBM samples. For TCGA-27-1 835, SEQ ID NO: 95 shows the reading frame at the breakpomt (bold), while SEQ ID NOS: 96-107, respectively, show alignments of the genomic breakpoints. For TCGA- 19-5958, SEQ ID NO: 108 shows the reading frame at the breakpomt (bold), while SEQ ID NOS: 109- I I 1 , respectively, show alignments of the genomic breakpoints. For TCG A-06-6390, SEQ ID NO: 1 12 shows the reading frame at the breakpoint (bold), while SEQ ID NOS: 113-131, respectively, show alignments of the genomic breakpoints. For TCGA- 12-0826, SEQ ID NO: 132 shows the reading frame at the breakpoint (bold), while SEQ ID NOS: 133-145, respectively, show alignments of the genomic breakpoints.
[0050] FIG. 2B shows recurrent gene fusions between FGFR and TACC genes in GBM. On the left, a gel of G ?-7¾CC-specific PGR is shown for FGFR3-TACC3 from a GBM cDNA sample. On the right, Sanger sequencing chromatograms show the reading frame at the breakpoint (SEQ ID N O: 81 ) and putative translation of the fusion protein (SEQ ID N O: 86) in the positive samples. [0051] FIG. 2€ shows recurrent gene fusions between FGFR and TACC genes in GBM. On the left, a gel of FGFR-TA C-specific PGR is shown for FGFR3-TA CCS from a GBM cDNA sample. On the right, Sanger sequencing chromatograms show the reading frame at the breakpoint (SEQ ID NO: 82) and putative translation of the fusion protein (SEQ ID NO:
87) in the positive samples.
[0052] FIG. 2D shows recurrent gene fusions between FGFR and TACC genes in GBM. Co-outlier expression of FGFR3 and TACC 3 in four GBM tumors from Atlas-TCGA is shown in the plot.
[0053] FIG. 2E shows recurrent gene fusions between FGFR and TACC genes in GBM. CNV analysis shows micro-amplifications of the rearranged portions of the FGFR3 and
TACC 3 genes in the same four Ailas-TCGA GBM samples,
[0054] FIG. 2F shows recurrent gene fusions between FGFR and TACC genes in GBM. On the left, a gel of FGFR-TA CC-specific PGR is shown for FGFR1-TACC1 from a GBM cDNA sample. On the right, Sanger sequencing chromatograms show the reading frame at the breakpoint (SEQ ID NO: 83) and putative translation of the fusion protein (SEQ ID NO:
88) in the positive samples.
[0055] FIG. 2G shows recurrent gene fusions between FGFR and TACC genes in GBM. On the left, a gel of FGFR-TA CC-specific PGR is shown for FGFR3-TACC3 from a GBM cDNA sample. On the right, Sanger sequencing chromatograms show the reading frame at the breakpoint (SEQ ID NO: 84) and putative translation of the fusion protein (SEQ ID NO:
89) in the positive samples.
[0056] FIG. 3A shows transforming activity of FGFR-TACC fusion proteins. FGFRl- TACC1 and FGFR3-TACC3 induce anchorage-independent growth in Rati A fibroblasts. The number of soft agar colonies was scored from triplicate samples 14 days after plating. Representative microphotographs are shown.
[0057] FIG. 3B are photomicrographs showing of immunofluoresence staining of tumors from mice injected with Ink4A;Arf-/~ astrocytes expressing FGFR3-TACC3 showing pGsidvity for glioma-specific (Nestin, Oig2 and GFAP) and proliferation markers (K167 and pHH3). Sub-cutaneous tumors were generated by Ink4A;Arf-/- astrocytes expressing FGFR- TACC fusions.
[0058] FIG. 3C shows Kaplan-Meier survival curves of mice injected intracranially with pTomo-shp53 (n = 8) or pTomo-EGFRvIIl-shp53 (n = 7) (green line; "light grey" in black and white image) and pTomo-FGFR3-TACC3-shp53 (n = 8, red line; "dark grey" in black and white image). Points on the curves indicate deaths (log-rank test, p = 0.0000 i, pTomo- shp53 vs. pTomo-FGFR3-TACC3-shp53).
[Θ059] FIG. 3D shows representative photomicrographs of Hematoxylin and Eosin staining of advanced FGFR3~TACC3~shp53 generated tumors showing histological features of high-grade glioma. Of note is the high degree of infiltration of the normal brain by the tumor cells. Immunofluorescence staining shows that glioma and stem cell markers (Nestin, OIig2 and GFAP), the proliferation markers (Ki67 and pHH3) and the FGFR3 -TACC3 protein are widely expressed in the FGFR3-TACC3-shp53 brain tumors. Fl -Tl : FGFR1- TACC1 ; F3-T3: FGFR.3-TACC3; F3-T3-K508M: FGFR3-TACC3-K508M.
[0060] FIG. 4A shows that FGFR3-TACC3 localizes to spindle poles, delays mitotic progression and induces chromosome segregation defects and aneuploidy Constitutive auto- phosphorylation of FGFR3-TACC3 fusion. Ink4A;Arf-/~ astrocytes transduced with empty lenfivirus or a lentivirus expressing FGFR3-TACC3 or FGFR3 -TACC3 -K508M were left untreated (0) or treated with 100 nM of the FGFR. inhibitor PD 173074 for the indicated times. Phospho-proteins and total proteins were analyzed by Western blot using the indicated antibodies.
[0061] FIG. 4B shows that FGFR3-TACC3 localizes to spindle poles, delays mitotic progression and induces chromosome segregation defects. Photomicrographs are shown of confocal microscopy analysis of FGFR3-TACC3 in Jnk4A;Arf-/~ astrocytes. Maximtm intensity projection of z-stacked images shows FGFR3-TACC3 (red; "dark grey" in black and white image) coating the spindle poles of a representative mitotic cell (upper panels), in telophase (lower panels) FGFR3-TACC3 localizes to the mid-body, a- tubulin (green; "grey" in black and white image), DNA (DAPI, blue; "light grey" in black and white image).
[0062] FIG. 4C shows representative fluorescence video-microscopy for cells transduced with vector or FGFR3-TACC3.
[0063] FIG. 4D shows a Box and Whisker plot representing the analysis of the time from nuclear envelope breakdown (NEB) to anaphase onset and from NEB to nuclear envelope reconstitution (TMER). The duration of mitosis was measured by following 50 mitoses for each condition by time-lapse microscopy.
[0064] FIG. 4E shows that FGFR3-TACC-3 localizes to spindle poles, delays mitotic progression and induces chromosome segregation defects. Representative images are shown of ceils with chromosome missegregation. Arrows point to chromosome misalignments, lagging chromosomes, and chromosome bridges.
[0065] FIG. 4F shows quantitative analysis of segregation defects in R ti A expressing FGFR1 -TACC1 and FGFR3-TACC3. F3-T3: FGFR3-TACC3; F3-T3-K508M: FGFR3- TACC3-K508M.
[0(566] FIG, 5A shows karyotype analysis of Rati A cells transduced with control, FGFR3, TACC3 or FGFR3-TACC3 expressing lentivirus. Distribution of chromosome counts of cells arrested in mitosis and analyzed for karyotypes using DAPL Chromosomes were counted in 100 metaphase cells for each condition to determine the ploidy and the diversity of chromosome counts within the cell population. FGFR3-TACC3 fusion induces aneuploidy.
[ΘΘ67] FIG, SB show's representative karyotypes and FIG. SC shows distribution of chromosome counts of human astrocytes transduced with control or FGFR3-TACC3 expressing lentivirus. Chromosomes were counted in 100 metaphase cells for each condition to determine the ploidy and the diversity of chromosome counts within the cell population.
[0068] FIG. 51) shows quantitati v e ana lysis of chromosome number in 100 metaphase ceils for each condition to determine the ploidy and the diversity of chromosome counts within the cell population. (n=3 independent experiments).
{0069'J FIG, 6A shows inhibition of FGFR-TK activity corrects the aneuploidy initiated by FGFR3-TACC3. The upper panel is a karyotype analysis of Rati A cells transduced with control or FGFR3-TACC3 lentivirus and treated with vehicle (DMSO) or PD173470 (100 liM) for five days. The lower panel shows the ploidy and the diversity of chromosome counts within the cell population were determined by quantitative analysis of chromosome number in 100 metaphase cells for each condition.
[0070] FIG. 6B shows inhibition of FGFR-TK activity corrects the aneuploidy initiated by FGFR3-TACC3. Correction of premature sister chromatid separation (PMSCS) by PD 173470 in cells expressing FGFR3-TACC3. Panels show representative metaphase spreads. DNA was stained by DAPL FIG. 6C shows quantitative analysis of metaphases with loss of sister chromatid cohesion (n=3; p = 0.001 , FGFR3-TACC3 treated with DMSO vs. FGFR3-TACC3 treated with PD 173470).
[0071] FIG. 7A shows inhibition of FGFR-TK activity suppresses tumor growth initiated by FGFR3-TACC3. Growth rate of Rat 3 A transduced with the indicated lentiviruses and treated for three days with increasing concentrations of PD173074. Ceil growth was determined by the MTT assay. Data are presented as the means±standard error (n-4).
10072] FIG. IB shows the growth rate of GSC- 1 123 treated with PD 173470 at the indicated concentrations for the indicated times. Cell grow th was determined by the MTT assay. Data are presented as the meansistandard error (n= ).
[0073] FIG . 7C shows the growth inhibitory effect of silencing FGFR3-TACC3 fusion. At the left, parallel cultures of GSC- 1 123 cells were transduced in triplicate. Rati A cells expressing FGFR3-TACC3 fusion were transduced with lentivirus expressing a non-targeting shRNA (Ctr) or shRNA sequences targeting FGFR3 (sh2, sh3, sh4). Five days after infection cells were plated at density of 2 104 cells/ well in triplicate and the number of trypan blue excluding cells was scored at the indicated times. Infection with lentivirus expressing sh-3 and sh-4, the most efficient FGFR3 silencing sequences reverted the growth rate of FGFR3- TACC3 expressing cultures to levels comparable to those of Rati A transduced with empty vector. Values are the means ± standard deviation (n = 3). At th right sided figure, GSC- 1 123 cells were transduced with lentivirus expressing a non-targeting shR A (sh-Ctr) or lentivirus expressing sh-3 and sh-4 sequences targeting FGFR3. estern Blot analysis was performed on parallel cultures using the FGFR3 antibody to the detect FGFT3-TACC3 fusion protein, β-actin is shown as a control for loading.
[0074] FIG, 7D shows that the FGFR inhibitor PD173074 suppresses tumor growth of glioma sub-cutaneous xenografts generated by Jnk4 A; ArjW- astrocytes expressing FGFR3- TACC3. After rumor establishment (200-300 mrnJ, arrow) mice were treated with vehicle or PD 173074 (50 rag/kg) for 14 days. Values are mean tumor volumes ± standard error (n = 7 mice per group).
[0075] FIG, 7E is a Kaplan- Meier analysis of glioma-bearing mice following orthotopic implantation of Ink4A;Arf-/- astrocytes transduced with FGFR3-TACC3. After tumor engraftment mice were treated with vehicle (n~9) or AZD4547 (50 mg/kg) (n~7) for 20 daysp = 0.001).
[0076] FIG. 8 shows a schematic of the TX-Fuse pipeline for the identification of fusion transcripts from RNA-Seq data generated from nine GSC cultures. The continued figure shows a schematic of the Exome -Fuse pipeline for the identification of gene fusion rearrangements fro DNA exome sequences of 84 GBM TCGA tumor samples.
[Θ077] FIG. 9 shows the validation of fusion transcripts iden tified by RNA-seq of nine GSCs. Sanger sequencing chromatograms show the reading frames at the breakpomt and putative translation of the fusion proteins in the positive samples (right side). The left side shows gels of RT-PCR conducted. (A) POLR2A- WRAP53. (B) CAPZB-UBR4. (C) ST8SIA4-PAM.. (D) P1GU- COA6.
[0078] FIG. E shows the fusion transcripts identified by whole transeriptome sequencing of nine GSCs. 54 split-reads are shown aligning on the breakpoint of the POLR2A-WRAP53 f sion. The predicted reading frame at the breakpoint is shown at the top with POLR2A sequences in red (left) and WRAP53 in blue (right). On the continued page, 48 spl it-reads are shown aligning on the breakpoint of the CAPZB-UBR4 fusion. The predicted reading frame at the breakpoint is shown at the top with CAPZB sequences in red (left) and UBR4 in blue (right). On the continued page after, 29 split-reads are shown aligning on the breakpoint of the ST8SIA4-PAM fusion. The predicted reading frame at the breakpoint is shown at the top with ST8SIA4 sequences in red (left) and PAM in blue (right). On the subsequent continued page, 17 split-reads are shown (top) aligning on the breakpoint of the PIGU-NCOA6 fusion. The predicted reading frame at the breakpoint is shown at the top with PiGU sequences in red (left) and NCO A6 in blue (right). Also (below), 6 split-reads are shown aligning on the breakpoint of the TF AR2-IL1 ORB fusion. The predicted reading frame at the breakpoint is shown at the top with IFNAR2 sequences in red (left) and IL10RB in blue (right).
[0(579] FIG, 10A shows the analysis and validation of the expression of fused transcripts in GSCs and GBM sample. Expression measured by read depth from RNA-seq data. Light grey arcs indicate predicted components of transcripts fused together. Overall read depth (blue; "grey" in black and white image) and split insert depth (red; "dark grey" in black and white image) are depicted in the graph, with a 50-read increment and a maximum range of 1800 reads. Note the very high level of expression in the regions of the genes implicated in the fusion events, particularly for FGFR3-TACC3.
[0(580] FIG. 10B shows the analysis and validation of the expression of fused transcripts in GSCs and GBM sample. Top panel, qRT-PCR showing the very high expression of FGFR3 and TACC3 mRNA sequences included in the FGFR3-TACC3 fusion transcript in GSC-1123. Bottom panel, for comparison the expression of sequences of WRAP53 mRNA included in the POLR2A-WRAP53 fusion in GSC-01 14 is also shown.
{0081} FIG. 0C shows the expression of the FGFR3-TACC3 protein in GSC-1 123 and GBM-1 123. Western blot analysis with a monoclonal antibody, which recognizes the N- ierminai region of human FGFR3 shows expression of a -150 kD protein in GSC-1 123 but not in the GSC cultures GSC-0331 and GSC-01 14, which lack the FGFR3-TACC3 rearrangement.
[0082] FIG. 10D shows the analysis and validation of the expression of fused transcripts in GSCs and GBM sample. Immunostaining analysis with the FGFR3 antibody of the tumor GBM-1 123 (top panel) and a GBM tumor lacking the FGFR3-TACC3 rearrangement.
FGFR3 (red; "light grey" in black and white image), DNA (DAPI, blue; "grey" in black and white image). The pictures were taken at low (left) and high (right) magnification.
[0083] FIG. WE shows MS/MS analysis of the -150 kD fusion protein
immunoprecipitated by the monoclonal anti-FGFR3 antibody from GSC-1123, identifying three unique peptides mapping to the FGFR3 (FGFR3 Peptide 1, 2, and 3) and three peptides mapping to the C-terminal region of TACC3 (TACC Peptide 1 , 2, and 3).
{0084'J FIGS. 11A-C shows Rati A cells transduced with control lentivirus or lentivurus expressing FGFR3, TACC3, FGFR3-TACC3 (FIG. 11 A) that were analyzed by Western blot with an antibody recognizing the N-terminus of FGFR3 (included in the FGFR3-TACC3 fusion protein) or the N-terminus of TACC3 (not included in the FGFR3-TACC3 fusion protein). FIG. 11B shows quantitative Western blot analysis of endogenous FGFR3-TACC3 in GSC-1 123 compared with lentivirally expressed FGFR3-TACC3 in RatlA. FIG. 11C shows Western blot analysis of FGFR3-TACC3 and FGFR3-TACC3-K508M in RatlA. a- !ubulin is shown as a control for loading.
[0085] FIGS. 11D-F shows expression analyses of FGFR3-TACC3 fusion construct (FIG. 11D) FGFR3 immunostaining of GBM- 1 123 (left, upper panel), BTSC 1 123 (right, upper panel), mouse GBM induced by FGFR3-TACC3 expressing lentivirus (left, lower panel), and sub-cutaneous xenograft of mouse astrocytes transformed by FGFR3-TACC3 fusion (right, lower panel); FGFR3-TACC3, red ("light grey" in black and white image); DNA (DAPI), blue ("grey" in black and white image). FIG. HE shows quantification of FGFR3-TACC3 positive cells in the tumors and cultures of cells shown in FIG. 11D. FIG. I IF shows a quantitative Western blot analysis of ectopic FGFR3-TACC3 fusion protein in mouse astrocytes and FGFR3-TACC3 induced mouse GBM (mGBM-15 and mGBM-17) compared with the endogenous expression in GBM1123. β-actin is shown as a control for loading. F3-T3: FGFR3-TACC3. a-tubulin or β-actin is shown as a control for loading. [0(586] FIG. 12A shows a western blot. Ink4A;Arf-/~ astrocytes transduced with empty lentivirus or a lentivirus expressing FGFR3-TACC3 were starved of mitogens and left untreated (time 0) or treated with FGF-2 at concentration of 50 ng/ml for the indicated times. Phospho-proteins and total proteins were analyzed by Western blot using the indicated antibodies, a-tubulin is shown as a control for loading.
{0087] FIGS. 12B show western blots. J.nk4A;Arf-/- astrocytes transduced with empty lentivirus or a lentivirus expressing FGFR3-TACC3 or FGFR3-TACC3-K508M were starved of mitogens and left untreated (time 0) or treated for 10 min with FGF~ 1 a the indicated concentrations. Phospho-proteins and total proteins were analyzed by Western blot using the indicated antibodies, β-actin is shown as a control for loading.
[0088] FIG, 12C show western blots. Ink4A;Arf-/- astrocytes transduced with empty lentivirus or a lentivirus expressing FGFR3-TACC3 or FGFR3 -TACC3 -K508M were starved of mitogens and left untreated (time 0) or treated for 10 min with FGF-8 at the indicated concentrations. Phospho-proteins and total proteins were analyzed by Western blot using the indicated antibodies, β-actin is shown as a control for loading.
[0089] FIGS. 12D-F shows mitotic localization of FGFR3-TACC3 fusion protein. FIG. 12 ) shows maximum intensity projection confocal image of a representative FGFR3-TACC3 expressing Ink4A;Arf-/- mouse astrocyte at metaphase irornunostained using the FGFR3 antibody (red; "dark grey" in black and white image), FGFR3-TACC3 displays asymmetric localization on top of one spindle pole. FIG. 12E shows max imum intensity projection confocal image of a representative TACC3 expressing Ink4A;Arf-/~ mouse astrocyte at metaphase immunostained with the TACC3 antibody (red; ("dark grey" in black and white image). TACC3 staining coincides with the spindle microtubules. FIG. 12F shows maximum intensity projection confocal image of a representative FGFR3 expressing Ink4A;Arj"-/- mouse astrocyte at metaphase immunostained with the FGFR3 antibody (red; ("dark grey" in black and white image). FGFR3 does not show a specific staining pattern in mitosis. Cells were co-immunosta ned using α-tubulin (green; ("light grey" in black and white image) to visualize the mitotic spindle. DNA was counterstained with DAPI (blue; ("grey" in black and white image). Images were acquired at 0.250 μηι intervals.
Endogenous levels of FGFR3 or TACC3 were undetectable under the applied experimental conditions. F3-T3: FGFR3-TACC3.
[0090] FIG, 13A shows sthat the FGFR3-TACC3 protein induces chromosomal mis- segregation, chromatid cohesion defects and defective spindle checkpoint. Quantitative analysis of meiaphase spreads for chromosome segregation defects in Ink4A;ARF-/~ astrocytes expressing vector control or FGFR3-TACC3 (upper panel). Microscope imaging analysis of chromosome segregation defects in Tnk4A;Arf-/~ astrocytes expressing FGFR3- TACC3 or vector control. Representative images of ceils with chromosome missegregation. Arrows point to chromosome misalignments, lagging chromosomes and chromosome bridges.
[0(591] FIGS. 13B-D shows representative images of premature sister chromatid separation (PMSCS) in Jnk4A;Arf-,/- astrocytes (FIG, 13B) and Rati A cells (FIG. 13C) expressing FGFR3-TACC3. Left, panels show representative metaphase spreads. Right, quantitative analysis of meiaphases with loss of sister chromatid cohesion. The number of mitosis with PMSCS in Ink4A;Arf-/- astrocytes was scored in at least 100 methaphases for each condition in three independent experiments. The number of mitosis with PMSCS was scored in triplicate samples of Rat i A cells. FIG. 13D is a graph showing nocodazole was added for the indicated durations to RatlA-H2B-GFP cells transduced with the specified lentiviruses. The mitotic index at each time point was determined by quantitating the H2B- GFP-positive cells in mitosis at each time point. Data are presented as average and standard deviation (n - 3). F3-T3: FGFR3-TACC3.
[0092] FIGS. 14A-B shows growth curves of human primary astrocytes transduced with lenti virus expressing FGFR3-TACC3 fusion or the empty vector. An analysis was conducted of FGFR3--TACC3 fusion mediated growth alteration and specific effect of RTK inhibitors on cells carrying FGFR-TACC fusions. FIG. 14A is a graph that shows cell proliferation of human primary astrocytes transduced with lenti virus expressing FGFR3-TACC3 fusion or the empty vector was determined by the MTT assay 7 days after infection (passage 1). Values are the means ± standard deviation (n = 4). />- value: 0.0033. FIG. 14B is a graph that shows cell proliferation of human primary astrocytes transduced with ientivirus expressing FGFR3- TACC3 fusion or the empty vector was determined by the MTT assay six weeks after the infection (passage 10). Values are the means±standard deviation (n=4). p-vaiue: 0.0018.
{0093'J FIGS. 14C-D shows specific growth inhibitory effect by FGFR inhibitors on FGFR-TACC fusion expressing cells. Cell growth was determined by MTT assay. Rati A cells transduced with the indicated Ientivirus were treated for three days with BGJ398 (FIG. 14C) or AZD4547 (FIG. 14D) at the indicated concentration. V alues are the means ± standard error (n = 4). [0(594] FIG, 14E shows the growth inhibitory effect of silencing FGFR3-TACC3 fusion, (left) GSC- i 123 cells were transduced in triplicate with ientivirus expressing a non-targeting shRNA (Ctr) or Ientivirus expressing sh-3 and sh-4 sequences targeting FGFR3, Five days after infection cells were plated at density of 2X104 ceils/well in triplicate and the number of trypan blue excluding cells was scored at the indicated times. Values are the means ± standard deviation (n = 3). (right) Western Blot analysis was performed on parallel cultures collected five days after infection using the FGFR3 antibody to the detect FGFT3-TACC3 fusion protein, β-actin is shown as a control for loading. (**: p-value = < 0.005: ***: -value = < 0.0001 ).
[0095] FIG, IS shows a survival plot of cells treated with PD 173074 , NVP-BGJ398, or AZD4547.
[0(596] FIG. 16 shows an FGFR3-TACC3 gene fusion identified by whole transcriptome sequencing of GSCs. The histogram describes the absolute frequency of each forward and reverse sequence read spanning the breakpoint.
[0097] FIG. 17 shows transforming activity of FGFR3 -TAG C 3. FGFR3 -TACC3 induces anchorage-independent growth in RatlA fibroblasts (top panels) and a transformed phenotype in Ink4A;Arf-/- primary astrocytes (bottom panels).
[0(598] FIG, 18 shows transforming activity of FGFR3-TACC3. Kaplan-Meier survival curves of mice injected intracranially with pTomo-shp53 (n = 8), pTomo-FGFR3-TACC3- shp53 (n - 8) and pTomo-EGFRvIII-shp53 (n = 7) are shown. Points on the curves indicate deaths (log-rank test, p = 0.025, pTomo-shp53 vs. pTomo-FGFR3-TACC3-shp53).
[0(599] FIG. 19 shows that inhibition of FGFR-TK activity corrects the aneupioidy and suppresses tumor growth initiated by FGFR3-TACC3. Short-term growth inhibition assays are shown of Ratl A. transduced with the indicated ientivirus and treated with PD 173470 at the indicated concentrations. Cells were treated for three days. Cell viability was determined by the MTT assay. Error bars show means ± standard error (n ------ 4).
[00100 j FIG, 20 is a growth inhibition assay of human astrocytes transduced with the indicated Ientivirus and treated for four days with PD 173470 at the indicated concentration. Cell viability was determined by the MTT assay. Error bars show means ± standard error (n = 4).
[Θ01Θ1] FIG. 21 is a graph showing a growth inhibition assay of human astrocytes transduced with the indicated lentivirus and treated for tour days with PD 173470 at the indicated concentration. Cell viability was determined by the MTT assay. Error bars show means ± standard error (n = 4).
[Θ0102] FIG. 22 shows graphs of the survival of Rati A cells in short-term growth inhibition assays, (Top graph) Rati A cells were transduced with the indicated ptomo constructs and treated with PD 173074 at the indicated concentrations . Cells were treated for three days. Cell viability was determined by the MTT assay. Error bars show means ± standard error (n - 4), In the bottom panel, a western blot photograph is shown,
[00103] FIG. 23 shows that inhibition of FGFR-TK activity corrects the aneupioidy and suppresses tumor growth initiated by FGFR3-TACC3. A plot is shown of karyotype analysis of Rat i A cells transduced with control or FGFR3-TACC3 lentivirus and treated with vehicle (DMSO) or PD173470 (100 nM) for five days.
[00164] FIG, 24 shows Survival of glioma-bearing mice was tracked following intracranial implantation of Ink4 A; Arf-/- astrocytes transduced with FGFR3-TACC3. After tumor engraftment mice were treated with vehicle or AZD4547 (50 nig/kg) for 20 days (vehicle, n = 7; AZD4547, n - 6; p - 0.0 1).
[00105] FIG. 25 shows the position of the peptides from FIG. 10E in the amino acid sequence of the FGFR3-TACC3 fusion protein, which are highlighted in pink (FGFR3; underlined) and blue (TACC3: dotted lines).
[00106] FIG. 26 shows Kaplan-Meier analysis ofIDH mutant and FGFR3-TACC3 positive human GBM. Log rank test p-value: 0,0169.
[00107] FIG. 27 is a picture that shows tumor xenografts that were induced following subcutaneous injection of J.nk4A;Arf-/~ mouse astrocytes transduced with lentivirus expressing FGFR3-TACC3 (upper panel A, right flank) or FGFRl-TACCl (lower panel B, right flank) fusion, but not with the empty vector (upper panel, left flank) or FGFR3-TACC3 carrying a K508M mutation in the kinase domain (FGFR3-TACC3-K508M; lower panel, left flank).
[!>Θ1Θ8] FIG. 28 shows constitutive auto-phosphorylation of FGFR3-TACC3 fusion. BTSC derived from FGFR3-TACC3 or RasV12 induced mouse GBM were left untreated or treated with 500nM PD173074 for the indicated times. Phospho-proteins and total proteins were analyzed by Western blot using the indicated antibodies, β-actin is shown as a control for loading. FIG. 29 shows Z-stacked confocal images of the representative FGFR3-TACC3 expressing lnk4A;Arf-/- mouse astrocyte shown as a maximum intensity projection. Cells were immunostained using FGFR3 (red; "dark grey" in black and white image) and a-tubulm (green; ("light grey" in black and white image). DNA was counter stained with DAP! (blue; ("grey" in black and white image). Images were acquired at 0.250 μηι intervals. Coordinates of the image series are indicated. F3-T3: FGFR3-TACC3.
[60110] FIG. 30 shows examples of SKY karyotype analysis painting two different cells from the same culture of GSC-1 123, illustrating the ongoing CIN and aneuploidy. Details of the karyotype analysis of 20 ceils are reported in Table 6.
[00111] FIG, 31 is a graphical representation of segmented CNVs data visualized using the Integrated Genomic Viewers software. Three bladder Urothelial Carcinoma harbor FGFR3-TACC3 gene fusions (black box). Red indicates amplification (A), blue indicates deletion (D).
80 12] FIG, 32 is a graphical representation of segmented CNVs data visualized using the Integrated Genomic Viewers software. One Breast Carcinoma harbors FGFR3-TACC3 gene fusions (black box). Red indicates amplification (A), blue indicates deletion (D).
[00113] FIG. 33 is a graphical representation of segmented CNVs data visualized using the Integrated Genomic Viewers software. One Colorectal Carcinoma harbors FGFR3 - TACC3 gene fusions (black box). Red indicates amplification (A), blue indicates deletion (D).
[0(5114] FIG. 34 is a graphical representation of segmented CNVs data visualized using the Integrated Genomic Viewers software. One Lung Squamous Cell Carcinoma harbors FGFR3-TACC3 gene fusions (black box). Red indicates amplification (A), blue indicates deletion (D).
[00115] FIG. 35 is a graphical representation of segmented CNVs data visualized using the Integrated Genomic Viewers software. One Head abd eck Squamous Cell Carcinoma harbors FGFR3 -TACC3 gene fusions (black box). Red indicates amplification (A), blue indicates deletion (D).
[00116] Glioblastoma multiformes (GBMs) are the most common form of brain tumors in adults accounting for 12-15% of intracranial tumors and 50-60% of primaiy brain tumors. GBM is among the most lethal forms of human cancer. The history of successful targeted therapy of cancer largely coincides with the inactivation of recurrent and oncogenic gene fusions in hematological malignancies and recently in some types of epithelial cancer. GBM is among the most lethal and incurable forms of human cancer. Targeted therapies against common genetic alterations in GBM have not changed the dismal clinical outcome of the disease, most likely because they have systematically failed to eradicate the truly addicting oncoprotein activities of GBM. Recurrent chromosomal rearrangements resulting in the creation of oncogenic gene fusions have not been found in GBM.
[©0117] GBM is among the most difficult forms of cancer to treat in humans (/). So far, the therapeutic approaches that have been tested against potentially important oncogenic targets in GBM have met limited success (2-4). Recurrent chromosomal translocations leading to production of oncogenic fusion proteins are viewed as initiating and addicting events in the pathogenesis of human cancer, thus providing the most desirable molecular targets for cancer therapy (5, 6). Recurrent and oncogenic gene fusions have not been found in GBM . Chromosomal rearrangements are hallmarks of hematological malignancies but recently they have also been uncovered in subsets of solid tumors (breast, prostate, lung and colorectal carcinoma) (7, 8). Important and successful targeted therapeutic interventions for patients whose tumors carry these rearrangements have stemmed from the discovery of functional gene fusions, especially when the translocations involve kmase-eoding genes (BCR-ABL, EML4-ALK) (9, 10).
[01)118] A hallmark of GBM is rampant chromosomal instability (CTN), which leads to aneuploidy ( 11). CIN and aneuploidy are early events in the pathogenesis of cancer (12). It has been suggested that genetic alterations targeting mitotic fidelity might be responsible for missegregation of chromosomes during mitosis, resulting in aneuploidy (13, 14).
[0(5119] Fibroblast growth factor receptors (FGFR) are transmembrane receptors that bind to members of the fibroblast growth factor family of proteins. The structure of the FGFRs consist of an extracellular ligand binding domain comprised of three Ig-like domains, a single transmembrane helix domain, and an intracellular domain with tyrosine kinase activity (Johnson, D.E., Williams, E. T. Structural and functional diversity in the FGF receptor multigene family. ( 1993) Adv. Cancer Res, 60: 1-41).
[00120] Transforming acidic coiled-coiled protein (TACC) stabilize microtubules during mitosis by recruiting mmispindles (Msps)/XMAP215 proteins to centrosomes. TACCs have been implicated in cancer.
[t>8121] From a medical perspective, the FGFR-TACC fusions provide the first "bona- fide" oncogenicaJly addictive gene fusions i GBM whose identification has long been overdue in this disease.
[t>8122] The practice of aspects of the present invention can employ, unless otherwise indicated, conventional techniques of ceil biology, ceil culture, molecular biology, transgenic biology, microbiology, recombinant DNA, and immunology, which are within the skill of the art. Such techniques are explained fully in the literature. See, e.g., Molecular Cloning A
Laboratory Manual. 3"" Ed., ed. by Sambrook (2001), Fritsch and Maniatis (Cold Spring Harbor Laboratory Press: 1989); DNA Cloning, Volumes 1 and II (D. N. Glover ed., 1985); Oligonucleotide Synthesis (M. J. Gait ed., 1984); Muilis et al. U.S. Pat. No: 4,683,195; jJcleje_Acid Hybridization (B. D. Hames & S. J. Higgins eds. 1984); Transcription and Transjlatiori (B. D. Hames & S. J. Higgins eds. 1984); Culture : Of .Aninral Cells (R. 1.
Freshney, Alan R. Liss, Inc., 1987); Immobilized Cells and Enzymes (IRL Press, 1986); B. Perbai, A Practical Guide To Molecular Cloning ( 1984); the series, Methods In Enzymology (Academic Press, Inc., N.Y,), specifically, Methods In Enzymology. Vols. 154 and 155 (Wu et al. eds.); Gene Transfer Vectors For Mammalian Cells (J, H, Miller and M. P. Caios eds., 1987, Cold Spring Harbor Laboratory); Immunochemical Methods In Cell And Molecular Biology. (Caner and Walker, eds., Academic Press, London, 1987); Handbook„Of
ExBerj ig tal (D. M. Weir and C. C. Blackwell, eds., 1986);
Manipulating the Mouse Embryo, (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1986). All patents, patent applications and references cited herein are incorporated by reference in their entireties.
[60123] One skilled in the art can obtain a protein in several ways, which include, but are not limited to, isolating the protein via biochemical means or expressing a nucleotide sequence encoding the protein of interest by genetic engineering methods.
[Θ0124] A protein is encoded by a nucleic acid (including, for example, genomic D A, complementary DNA (cDNA), synthetic DNA, as well as any form of corresponding RNA). For example, it can be encoded by a recombinant nucleic acid of a gene. The proteins of the invention cars be obtained from various sources and can be produced according to various techniques known in the art. For example, a nucleic acid that encodes a protein can be obtained by screening DNA libraries, or by amplification from a natural source. A protein can be a fragment or portion thereof. The nucleic ac ds encoding a protein can be produced via recombinant DNA technology and such recombinant nucleic acids can be prepared by conventional techniques, including chemical synthesis, genetic engineering, enzymatic techniques, or a combination thereof. For example, a fusion protein of the invention comprises a tyrosine kinase domain of an FGFR protein fused to a polypeptide that constitutively activates the tyrosine kinase domain of the FGFR protein. For example, a fusion protein of the invention comprises a transforming acidic coiled-coil (TACC) domain fused to a polypeptide with a tyrosine kinase domain, wherein the TACC domain constitutively activates the tyrosine kinase domain. An example of a FGFR1 -TACC 1 polypeptide has the amino acid sequence shown in SEQ ID NO: 150. An example of a FGFR3-TACC3 protein is the polypeptide encoded by the nucleic acid having the nucleotide sequence shown in SEQ ID NO: 94, Examples of a FOFR3-TACC3 polypeptide has the amino acid sequence shown in SEQ ID NO: 79, 158, 159, 160, or 161.
[Θ0125] The Genbank ID for the FGFR3 gene is 2261. Three isoforms are listed for FGFGR3, e.g., having Genebank Accession Nos. NP 000133 (corresponding nucleotide sequence NM_000142); NP_001156685 (corresponding nucleotide sequence
NM 001 163213); NP 075254 (corresponding nucleotide sequence NM 022965).
|0O126j SEQ ID NO: 90 is the FGFR3 Amino Acid Sequence, Transcript Variant 1
000133; 806 aa).
1 MGAPACALAL CVAVAIVAGA SSESLGTEQR WGRAAEVPG PSPGQQEQLV FGSGDAVELS 61 CPPPGGGPMG PTVWVKDGTG LVPSERVLVG PQRLQv'LNAS HEDSGAYSCR QRLTQRVLCH 1 21 FSVRVTDAPS SGDDEDGEDE AEDTGVDTGA PYBTRPERMD KKLLAVPAAN TVRFRCPAAG 181 NPTPSIS LK NGREFRGEHR XGGIKLRHQQ WSLV ESWP SDRGNYTC ENKFGS IRQT 24 1 YTLDVLERSP HRPILQAGLP ANQTAVLGSD VEFHCKVYSD AQPHIQwLKH VEVNGSKVGP 301 DGTPYVTVLK TAGANTTDKE LEVLSLHNVT FEDAGEYTCL AGNSIGFSHH SAWL LPAE
3 61 EELv'EADEAG SVYAG ILS YG VGFFLFXL AAVTLCRLRS PPKKGLGSPT VHKISRFPLK
4 21 RQVSLESKAS MSSKTPLVRI ARLSSGEGPT LAKVSELELP ADPKWELSRA RLTLGKPLGE 4 81 GCFGQWMAE AIGIDKDRAA KPVTVAVKML KDDA DK DL S DLVSEMEMMK IGKHKN IIN 54 1 LLG.ACTQGGP LYVLVEYAAK GNLREFLRAR RPPGLDYSFD TCKPPEEQLT FKDLVSCAYQ 601 VARGMEYLAS QKC I HRDLAA RNVLVTEDNV KIADFGLAR DVH LDYYKK TTNGRLPVK 661. MAPEALFDRV ? H Q S D V W S F GVLL EXFTL GGSPYPGIPV ESLFKLLKSG H R DK P A C 721 HDLYMIMREC WHAAPSQRPT F QLVSDLDR VL VTSTDEY LDLSAPFEQY 3PGGQDTPSS 781 SSSGDDSVFA RDLLPPAPPS SGGSRT
| 0 ! 27] SEQ ID NO: 91 is the FGFR3 Nucleotide Sequence, Transcript Variant 1 (NM 000142; 4304 bp). i gtcgcgggca gctgqcqccq cgcggtcctg ctctqccqqt cacacgqacq caccggcgqq
61 ccgccggccg gagqgacggg gcgggagctg ggcccgcgga cagcgagccg gagcgggagc 121 cgcgcgtagc qagccqqqct ccqqcgctcq ccagtctccc gagcggcgcc cgcctcccgc 181 cggtc!cccgc gccgqqccg t gggggqcagc a gcccc!ccic gcgc gcctg aqqacgccgc 241 ggcccccgcc cccgccatgg gcgcccctqc tgcgccctc gcgctc qcq tggccgtggc 301 catcgtggcc ggcgcctcct cgaagtcctt ggqqacggag caqcqcqtcq tqqqqcqaqc
361 ggcagaagtc ccqggcccacj agcccqgcca cjcaggagcag ttggtcttcg qcaqcgcjcjga 421 gctgtggag ctgagctgtc ccccgcccgq gggtggtccc atggggccca ctgtctgggt
481 caaggatggc acagggctgg tgccctcgga gcgtgtcctg gtggggcccc aqcggc gca 541 ggtgctgaat gcctcccacg agqactccqq qqcctacagc tqccqqcaqc gac cacgca 601 gcgcgtactg tqccacttca qtqtgcgqqt gacagacgct ccatcctcgq qaqatqacqa 661 agacgcjggag gacgaggctc; aggacacagg tcjtggacaca ggggcccctt actggacacg 721 gcccgagcgg atggacaaga agctgc ggc cgtgccggcc gccaacaccg tccgcttccg 781 c gcccagcc gctggcaacc ccactccctc catctcc qq ctgaagaacg qcagggagtt 841 ccgcggcgag caccqcattq gagqcatcaa qctgcgqcat cagcagtgga gcctgqtcat 901 ggaaagcgtg gtgccctcgg accgcqqcaa ctacacctqc qtc qqaaa acaaatttqq 961 cagcatccgg cagacgtaca cgctcjqacgt gctggagcgc tccccgcacc ggcccatcct
1021 gcaggcgggg ctgccqqcca accagacggc ggtgctgggc aqcgacgtqg agttccactg 1081 caaggfcgtac agtgacgcac agccccacat ccagtggctc aagcacgtgg aqqtgaa gq 1141 cagcaaggtg ggcccggacg gcacacccta cqttaccqtg ctcaagacgq cgggcgctaa 1201 caccaccgac aaggacjctag aggttctctc cttcjcacaac gtcacctttg aggacgccgg 1261 ggagtacacc tgcctggcgg gcaattctat tgggttttct catcactctg cgtggctggt 1321 ggtgctgcca gccgagqagg aqctggtgga ggctgacgag gcgggcagtg tgtatgcagg 1381 catcc caqc tacggggtgg gcttcttcct g ttcatcctg gtgg gqcgg ctgtgacgct 1441 ctgccgcctg cgcagccccc ccaagaaaqq cctgggctcc cccaccg gc acaagatctc 1501 CCCjCttCCCCj ctcaagcgac aggtgtccct ggagtccaac cjcgtccatqa gctccaacac 1561 accactggtg cgcatcgcaa ggc gtcctc aggggagggc cccacgctgg ccaatgtctc 1621 cgagctcgag ctgcctqccg accccaaatq ggagctgtct cgggcccggc tgaccctggg 1681 caagcccctt ggqgagggct qcttcqqcca ggtggtcatg gcggagqcca tcggca ga 1741 caaggaccgg gccgccaagc ctgtcaccgt aqccqtqaag atgctgaaag acqatqccac 1801 tcjacaaggac ctgtcggacc tggtgtctga gatcjcjagatg atgaagatqa tccjcjqaaaca 1861 caaaaacatc atcaacctgc tgggcqcctg cacgcaggqc gggcccctgt acgtgctggt 1921 ggagtacgcg gccaagqgta acctgcggga gtttctgcgg gcgcggcggc ccccgggcct 1981 ggactactcc ttcgacacct gcaaciccgcc cgaggagcag ctcaccttca aqqacc qqt 2041 gtcctgtgcc taccaqqtqq cccgqggcat qqaq acttq qcctcccaqa aqt catcca 2101 cacjggacctcj gctgcccgca atqtgctcjcjt gacccjaggac aacgtgatga agatcqcaga 2161 cttcgggctg gcccgggacg tgcacaacct cqactactac aagaaga aa ccaacggccg 2221 gc gcccg g aagtggatgg cgcctgaggc cttg ttgac cqaqtc aca ctcaccagag 2281 tgacqtctqq tcctttgggg tcctgctctg ggaqatcttc acgc ggggg gctccccqta 2341 ccccggcatc cctgtqqaqq agctcttcaa qctqctqaaq qaqqqccacc qcatqqacaa 2401 gcccgccaac tgcacacacg acctgtacat gatcatgcgg qagtgcfcggc atgccgcgcc 2461 cteccagagg cccaccttca agcagctggt ggaggacctg gaccgtgtcc ttaccgtgac 2521 gtccaecgac gaqtacctqq acctgtcggc gcctttcqaq caqtactccc cgggtggcca 2581 ggacaccccc agctccagct cctcaqqqqa cqactccqtq tttqcccacq acctgctgcc 2641 cccggcccca cccagcagtg ggggctcgcg gacgtgaagg qccactgqtc cccaacaatg 2701 tgaggggtcc ctagcagccc accctgctcjc tggtgcacag ccactccccg gcatqagact 2761 cagtgcagat ggagagacag ctacacagag ctttggtctg tgtgtgtgtg tqtgcgtgtg 2321 tg tgtgtgfcg tqtgcacatc cgcgtgtgcc tgtg tgcgtg cqcatc qc ctccagqtgc 2881 agaggtaccc tggqtqtccc cgctgctgtg caacgatctc ctqactqqtq ctgcaqcacc 2941 gaggggcctt tcjttctggcjcj ggacccagtg cagaatgtaa gtgggcccac ccgqtgcjcjac 3001 ccccgtggcjc; cagggagctg cjcjcccgacat ggctccggcc tctgcctttg caccacggga 3061 catcacaggg tgggcctcgg cccctcccac acccaaagct gagcctgcag qqaagcccca 3121 ca qtccagc accttqtqcc tgqqqtgtta gtggcaccgc ctccccacct ccagqctttc 3181 ccacttccca ccctqcccc cagagactga aattacgggt acctgaaqat qqqaqccttt 3241 accttttatg caaaaggttt attccqgaaa ctagtgtaca tttctataaa tagatgctgt 3301 gtatatggta tata acata tatatatata acatatatgg aagaggaaaa ggctq taca 3361 acggaggcct gcgaccctgg gggcacagga ggcaggcatg gccctgggcg qqgcgtgggg 3421 gggcgtggag ggaggcccca ggqqqtc ca cccatgcaag cagaggacca gggccttttc 3481 tggcaccgca gttttgtttt aaaactggac ctgtatattt gta.aagctat ttatgggccc 3541 ctggcac ct tgt cccaca ccccaacact tccagcattt agctggccac atggcggaga 3601 gttt aattt aactta t gacaaccgag aaggtt atc ccgccgatag agggacggcc 3661 aagaatgtac gtccagcctg ccccggagct ggaggatccc ctccaagcct aaaaggttg 3721 taatagttgg aggtgattcc agtgaagata ttttatttcc tttgtccttt ttcaggagaa 3781 ttagatttct ataggatttt tctttaggag atttattttt tggacttcaa agcaagctgg 3841 attttca a caaa tcttc aattgc gt gtgtcccagg cagggagacg gtttccaggg 3901 aggggccggc cctgtgtgca ggttccgatg tattagatg ttacaagttt atatatatct 3961 atatatataa tttattgagt ttttacaaga tgtatttgtt gtagact aa cacttcttac 4021 gcaatgcttc tagagtttta tagcctggac tgctaccttt caaagcttgg agggaagccg 4081 tgaat cagt tcjgttcgttc tgta.ctgtta ctgggccctg agtctgggca gctgtccctt 4141 gcttgcc gc agggccatgg ctcagggtgg tctcttctt.g gggcccagtg ca ggtggcc 4201 agagg tgtca cccaaaccgg caggfcgcgat tttgttaacc cagcgacgaa ctttccgaaa 4261 aataaagaca cctgg ttgct aacctggaaa aaaa
The Genbank ID for the TACC3 gene is 10460. SEQ ID NO: 92 is the TACC3 Amino Acid Sequence (NP 006333) (838 aa).
1 MSLQVL DKN VSNEKNTENC DFLFSPPBVT GRSSVLRVSQ KKNVPPKNLA KAMKv'TFQTP
61 LRDPQTHRIL SPSMASKLEA PFTQDDTLGL E SHPVWTQK ENQQLIKEVD AKTTHG1LQK
121 PVEADTDLLG DASPAFGSGS SSESGPGALA DLDC3SSSQS PGSSE QMVS PGKVSGSPEQ
181 AVESKLSSYS LDRRV ΡΑΞΕ TLSDPCRTES QHKAETPHGA ESECKAETPH GAEESCRHGG
241 VCAP.AA.vATS PPGAIPKEAC GGAPLQGLPG EALGCPAGv'G TPVPA.DGTQT LTCAHTSAPE
301 S A.P?. HLVA GRAMTLSPQE EVAAGQMASS SRSGPVKLEF DVSDGATSKR APPPRRLGER
361 SGLKPPLRKA AVRQQKAPQE VEΞDDGRSGA GEDPPMPASR GSYHLDWDKM DDPNFIPFGG 421 DTKSGCSEAQ PPESPETRLG QPAAEQLHAG PATEEPGPCL 3QQLHSA3AE DTPWQLAAE
481 ΤΡΤΑΞΞΚΞΡΑ LNSASTSLPT SCPGSSPVPT HQQGQPALSL KSSSFRDPAS VLGTGAEVDY 541 LEQFGTSSFK ESALRKQSLY LKFDPLLRDS PGRPVPVATE TSSMHGA ET PSGRPREAKL 601 VEFDFLGALD IPVPGPPPGV PAPGGPPLST GPIVDLLQYΞ QKDLD VVK& TQEE RELRS 661 RCEELHGK L ELGKI DRFE EWYQAMEEV QKQKELSKAE IQKVLKEKDQ LTTDLNSMEK 721 SFSDLFKRFE KQKEVIEGYR K EESLKKCV EDYLARITQE GQRYQALKAH AEEKLQLANE 781 EIAQVRSKAQ AEALALQASL RKSQMRIQSL EKTVEQKTKE NEELTRICDD LISKMEKI
[110129] SEQ ID NO: 93 is the TACC3 Nucleotide Sequence (NM 006342) (2847 bp): i gcgtttgaaa ctccggcgcg ccggcggcca tcaaggcjcta gaagegegae ggeggtagea
61 gctaggct g gcccccggcg ggagcagac gcggacccc ccttcctggc ggcggcggcg 121 cgggc caga gcccggcaac gggcgggcgg gcagaatgag tetgeaggtc ttaaacgaca 181 aaaatgtcag caatgaaaaa aatacagaaa attgcgactt cctgtt tcg ccaccagaag 241 ttaccggaac; atcgtctgtt cttcgtgtgt cacacjaaaga aaatgtcjcca cccaagaacc 301 tggccaaagc tatgaaggtc; acttttcaga cacctctgcg ggatccacag acgcacacjga 361 t ctaagtcc tagcatggcc agcaaacttg aggctcctt cactcaggat gacacccttg 421 gactggaaaa c cacacccg gtctggacac agaaagagaa ccaacagctc atcaaggaag 481 tggatgccaa aactactcat ggaattctac agaaaccagt ggaggctgac accgacctcc 541 tgggggatgc aagcccagcc tttgggagtg gcagctccag cgag ctggc ccaggtgccc 601 tggctcjacct ggactgctca agctcttccc acjagcccagg aagttctgag aaccaaatgg 661 tgtctccagg aaaagtgtct ggcagccctg ageaageca ggaggaaaac cttagttcct 721 attcc taga cagaagag g acacccgcct ctgagaccct agaagaccct tgcaggacag 781 ag cccagca caaagcggag actccgcacg gagecgagga agaatgeaaa geggagaetc 84 ccjcacggacjc cgaggaggaa tcjccggcacg gtggqgtctg tgctcccgca gcagtggcca 901 cttcgcctcc tcjgtgcaatc cctaaggaag ectgeggagg agcacccctg cagggtctgc 961 ctggcgaagc cctgggctgc cctgcgggtg tgggcacccc cgtgccagca gatggcactc 1021 agacccttac c gtgcacac acctctgctc ctgagagcac agccccaacc aaccacctgg 1081 tggctggcag ggccatgacc ctgagtcctc aggaagaagt ggctgeagge caaatggeca 114 gctcctcgag gagcggacct cjtaaaactag aatttgatgt atctgatggc gccaccagca 1201 aaagggcacc cccaccaagg agac gggag agaggtcegg cctcaagcct ccctt.gagga 1261 aagcagcagt gaggcagcaa aaggccccgc aggaggtgga ggaggacgac ggtaggagcg 1321 gagcacjgaga ggaccccccc atgccagctt ctcggggctc tta.ccacctc gactgggaca 1381 aaatgga ga cccaaacttc atcccgttcg gaggtgacac caagtc ggt tgcagtgagg 1441 cccagccccc agaaagccct gagaccaggc tgggccagcc agcggctgaa cagttgcatg 1501 ctgggcctgc cacggaggag ccaggtccct gtctgagcca gcagctgcat tcagcctcag 1561 cggaggacac gcctgtggtg cagttggcag ccgagacccc aacagcagag agcaaggaga 1621 gagccttgaa ctctgccacjc acctcgcttc ccacaagctg tccacjgcagt gagccagtgc 1681 ccaccca ca gcaggggcag cctgcc gg agctgaaaga ggagagcttc agagaccccg 1741 ctgaggttct aggcacgggc gcggaggtgg abtacctgga gcag bttgga acttcctcgt 1801 ttaaggagtc ggccttgagg aagcagtcct tatacctcaa gttcgacccc. ctcctgaggg 1861 acagtcctgg tagaccagtg cecgtggcea ccgagaccag cagcatgcac ggtgcaaatg 1921 agactccctc acjgacgtcccj cgggaagcca agcttgtgga gttcgatttc ttgggacjcac 1981 ggacat cc tgtgccaggc ccacccccag gtg tcccgc gcctgggggc ccacccctgt 2041 ccaccggacc atagtggac ctgctccagt acagccagaa ggacctgga gcagtggtaa 2101 aggcgacaca ggaggagaac cgggagctga ggagcaggtg tgaggagctc cacgggaaga 2161 acctggaact ggggaagatc atggacaggt tcgaagaggt tgtgtaccag gccatggagg 2221 aagttcagaa gcagaaggaa ctttccaaag ctgaaatcca gaaagttcta aaagaaaaag 2281 accaact ac cacagatctg aactccatgg agaagtcct ctccgacctc ttcaagcg t 2341 ttgagaaaca gaaagagg tg atcgagggct accgcaagaa cgaagagtca ctgaagaagt 2401 gcgtggagga ttacctggca aggatcaccc aggagggcca gaggtaccaa gccctgaagg 2461 cccacgcgga ggagaagctg cagctggcaa acgaggagat cgcccaggtc cggagcaagg 2521 cccaggcgga acjcgttggcc ctccaggcca gcctgaggaa ggagcagatg cgcatccagt 2581 cgctggagaa gacagtggag cagaagacta aagagaacga ggagc gacc aggatctgcg 2641 acgacctcat cfcccaagafcg gagaagatct gacctccacg gagccgctgt ccccgccccc 2701 ctgctcccgt ctgtctgtcc tgtctgattc tcttaggtgt catgttcttt tttctgtctt 2761 gtcttcaact tttt aaaaa ctagattcjct ttgaaaacat gactcaataa aacjtttcctt 2821 tcaatttaaa cactgaaaaa aaaaaaa ] SEQ ID NO: 94 is the nucleotide sequence of FOFR3-TACC3. i gtcgcgggca gctggcgccg cgcggtcctg ctc gccgg cgcacggacg caccggcggg
61 gccgg g gagggacggg gcgggagctg ggcccgcgga cagcgagccg gagegggage 121 cgcgcgtagc gagccgggct ccggcgctcg ccagbctccc gagcggcgcc cgcctcccgc 181 ccjcjtgccccjc gccgggccgt ggggggcagc atgcccgcgc cjcgctgcctg aggacgccgc 241 ggcccccgcc cccgcca gg gcgcccctgc c gcgccctc gcgc ctgcg tggccgtggc 301 catcgtggcc ggcgcctcct cggagtcctt ggggacggag cagcgcg cg tggggegage
361 ggcagaagtc ccgggcccag agcccggcca gcaggagcag ttgg betteg geagegggga 421 tgctgtggag ctgagctgtc ccccgcccgg gggtggtccc atggggccca ctgtctgggt
481 caaggatgcjc acagggctgg tgccctcgga gcgtgtcctg gtggggcccc ageggctgea 541 ggtgctgaat gcctcccacg aggactccgg ggcctacagc tgccggcagc ggctcacgca 601 gcgcgtacfcg tgccacttca g gtgcgggt gacagacgct ccatccbcgg gagatgacga 661 agacggggag gacgaggctg aggacacagg gtggacaca ggggcccctt actggacacg 721 gcccgagcgg atggacaaga agctgctggc cgtgccggcc gccaacaccg tccgcttccg 781 ctgcccagcc gctgcjcaacc ccactccctc catctcctgg ctgaagaacg gcagggagtt 841 ccgcggcgag caccgcattg gaggcatcaa gctgcggcat cagcagtgga gectggbcat 901 ggaaagcgtg gtgccctcgg accgcggcaa ctacacctgc gtcgtggaga acaagtttgg 961 cagcatccgg cagacgtaca cgctggacgt gctggagcgc tccccgcacc ggcccabcct
1021 gcaggcgggg ctgccggcca accagacggc ggtgctgggc agcgacgtgg agttccactg 1081 caaggtgtac agtgacgcac acjcccca.cat cca.cjtggctc aagcaccjtgg agcjtgaatgcj 1141 cagcaaggtg ggcccggacg gcacacccta cgttaccgtg etcaagaegg egggegctaa 1201 caccaccgac aaggagctag aggttctcbc cttgcacaac gtcaccbbtg aggaegcegg 1261 ggagtacacc tgcctggcgg gcaattctat tgggttttct catcactctg cgtggctggt 1321 ggtgctgcca gccgaggagg agctggtgga ggctgacgag gcgggcagtg tgtatgeagg 1381 catcctcacjc tacggggtgg cjcttcttcct gttcatcctg gtggtggcgg ctgtgacgct 1441 ctgccgcctg cgcagccccc ccaagaaagg cctgggc cc cocaccgtgc acaagabctc 1501 ccgcttcccg ctcaagcgac aggtgtccct ggag bccaac gcgtccabga gcbccaacac 1561 accactggtg cgcatcgcaa ggctgtcctc aggggagggc cccacgctgg ccaatgtctc 1621 cgagctcgag ctgcctgccg accccaaabg ggagctgtct cgggcccggc tgaccctggg 1681 caagcccctt ggggagggct cjcttcggcca ggtggtcatg cjcggagcjcca teggcattga 1741 caaggaccgg gccgc aagc ctg caccgt agccgtgaag atgctgaaag acgatgccac 1801 tgacaaggac ctgtcggacc bggtgtc ga gatggagatg atgaagabga tegggaaaca 1861 caaaaacatc atcaacctgc tgggcgcctg cacgcagggc gggcccctgt acgtgctggt 1921 ggagtacgcg gccaagggta acctgcggga gtttctgcgg gcgcggcggc ccccgggcct 1981 ggactactcc ttcgacacct gcaagccgcc cgaggagcag ctcaccttca aggacctggt 2041 gfccctgtgcc taccaggtgg cccggggcat ggagtacttg gcctcccaga agfcgcatcca 2101 cagggacctg gctgcccgca atgtgctggt gaccgaggac aacgtgatga agatcgcaga 2161 cttcgggctg gcccgggacg tgcacaacct cgactactac aagaagacaa ccaacggccg 22.2.1 gctgcccgtg aagtggatgg cgcctgaggc cttgtttgac cgagtctaca ctcaccagag 2281 tgacgfcctgg tcctttgggg tcctgctctg ggagatcfctc acgc ggggg gctccccgta 2341 ccccggcatc cctgtggagg agctcttcaa gctgctgaag gagggccacc gcatggacaa 2401 gcccgccaac tgcacacacg acctgtacat gatcatgcgg gagtgctggc atgccgcgcc 2461 ctcecagagg cccaccttca agcagctggt ggaggacctg gaccgtgtcc ttaccgtgac 2521 gtccaccgac tttaaggagt cggccttgag gaagcagtec ttatacctca agttcgaecc 2581 cctcctgagg gacagtcctg gtagaccagt gcccgtggcc accgagacca gcagcatgca 2641 cggtgcaaat gagactccct caggacgtcc gcgggaagcc aagcttgtgg agttcgattt 2701 cttgggagca ctggacattc ctgtgccagg cccaccccca ggtgttcccg cgcctggggg 2761 cccacccctg tccaccggac ctatagtgga cctgctccag tacagccaga aggacctgga 2821 gcagtggta aaggcgacac aggaggagaa ccgggagctg aggagcaggt gtgaggagct 2881 ccacgggaag aacctggaac tggggaagat catggacagg ttcgaagagg ttgtgtacca 2941 ggccatggag gaagttcaga agcagaagga actttccaaa gctgaaatcc agaaagttct 3001. aaaagaaaaa gaccaactta ccacagatct gaactccatg gagaagtcct tctccgacct 3061 cttcaagcgt tttgagaaac agaaagaggt gatcgagggc taccgcaaga acgaagagtc 312.1 actgaagaag tgcgtggagg attacc ggc aaggatcacc caggagggcc agaggtacca 3181 agccckgaag gcccacgcgg aggagaagct gcagctggca aacgaggaga tcgcccaggt 3241 ccggagcaag gcccaggcgg aagcgttggc cctccaggcc agcctgagga aggagcagat 3301. gcgcatccag tcgctggaga agacagtgga gcagaagact aaagagaacg aggagctgac 3361 caggatctgc gacgacc ca tctccaagat ggagaagatc tgacctccac ggagccgctg 342.1 tccccgcccc cctgctcccg ctgt.c gtc ctgtctgatt ctct.taggtg tcatgtt.c t 3481 ttttcfcgtct tgtcttcaac ttttfctaaaa actagatfcgc tttgaaaaca tgactcaata 3541 aaagtttcct ttcaatttaa acactgaaaa aaaaaaaa
[110131] The Genbank ID for the FGFRl gene is 2260. Eight isoforms are listed for
FGFRl, e.g., having Genebank Accession Nos. NP_001167534 (corresponding nucleotide sequence NM 00 I I 74063); NP 001 167535 (corresponding nucleotide sequence
NM 001 174064): NP 001 167536 (corresponding nucleotide sequence NM 001 174065 ); NP_001167537 (corresponding nucleotide sequence NMJ301 174066): NP_001167538
(corresponding nucleotide sequence NM_001 1 74067); NP_056934 (corresponding nucleotide sequence NM 015850); NP ...075593 (corresponding nucleotide sequence NM . 23105); NP 075594 (corresponding nucleotide sequence NM 023106); NP 075598 (corresponding nucleotide sequence NM__0231 10).
[00132] SEQ ID NO: 146 is the FGFR 1 Amino Acid Sequence for isoform 10, having Genebank Accession No. NP_0G1167534 (820 aa):
1 M SWKCLLF AVLVTATLCT ARPSPTLFEQ AQPWGAPVEV ESFLVHPGDL LQLRCP.LRDD
61 VQSI WLRDG VQLAES RTR ITGEEVEVQD SVPADSGLYA CVTSSPSGSD TTYFSVNVSD
121 ALPSSEDDDD DDDSSSEEKE TD TKPKRMP VAPYWTSPEK MEKKLR VPA AKTVKFKCPS
181. SGTPNPTLRW LKNGKEFKPD HRIGGYKVRY ATWSIIMDSV VPSDKG YTC IVENEYGΞI
241 RTYQLD ER SPHRPILQAG LPANKTVALG SNVEFMCKVY SDPQPRIQVJL KHIEVN6SKI
301 GPDNLPYVQI LKTAGVNTTD KEMEVLHLRN VSFEDAGEYT CLAGNSIGLS HHSAWLTVLE
361 ALEERPAV T SPLYLEIIIY CTGAFLISCM VGSVIVYKMK SGTKKSDFHS QtiAVHKLAKS
421 I LRRQv'SAD SSASMNSGVL LVRPSRLSSS GTP LAGVSE YELPEDPRWE LPRDRLVLGK 481 PLGEGCFGQV VLAEAIGLDK DKP Rv'TKVA VKMLKSDATE KDLSDLISEM EMMKMIGKHK 541 IINLLGA QDGPLYVIVE YASKGNLREY LQARRPPGLE YCY PSH PE EQLSSKDLVS 601 CAYQVARGME YLASKKCIHR DLAARNVLVT EDKVMKIADF GLARDIHHID YYKKTT GRL 661 PVKWMAPEAL FDRIYTHQSD V SFGVLL E IFTLGGSPYP GVPVEELFKL LKEGHRMDKP 721 S CTKELYM MRDCWHAVPS QRPTFKQLVE DLDRIVALTS NQEYLDLSMP LDQYSPSFPD 781 TRSSTCSSGE DSVFSHEPLP EEPCLPRHPA QLA.' GGLKRK
[00133] SEQ ID NO: 147 is the FGFR l Nucleotide Sequence for isoform 10, having Genebank Accession No. NM_0G1 174063 (5895 bp):
1 agatgcaggg gcgcaaacgc caaaggagac caggctgtag gaagagaagg gcagagcgcc
61 ggacagctcg gcccgctccc cgtcctttgg ggccgcggct ggggaactac aaggcccagc 121 aggcagctgc aqggggcgga ggegejaggag ggaccagcgc gggtgggagt qagagacjega 181 gccctcgccjc cccgccggcg catagcgctc ggagegctet tgcggccaca ggcgcggcgt 241 cctcggcggc ggqeggcage tagegggage cgggacgccg gtgcagccgc aqcgcgcgga 301 ggaacccggg tgtgccggga getgggegge eacg teegga cqggaccgag acccctcgta 361 gcgcattgcg gcgacctcgc cttccccggc cgcgagcgcg ccgc gcttg aaaagccgcg 421 gaacccaagg acttttctcc ggtccgagct cggggcgccc cgcagggcgc acggtacccg 481 tcjctgcagtc gggcacgccg cggcgccggg gcctccgcag gqcgatgqag cccggtctgc 541 aaggaaagtg aggcgccgcc gctgcqttct ggaggaggqg ggcacaagg ctggagaccc 601 cgggtggcgg e gggag c ccceccgcc ecc!cetcegg gqcaccagct ccggctcca 661 tgttcccgcc egggctggag gcgccgagca ccgagcgccg cegggagteg agcgccggcc 721 gcggagctct tcjcgaccccc; ccaggacccg aacagagccc gggggcggcg ggccggagcc 781 gggqacgcgg gcacacqccc gctcgcacaa gccacqgcgg actctcccqa ggeggaaect 841 ccacgccgag cgagggtcag tttgaaaagg aggatcgaqc tcactgtgga qtatccabgg 901 agatgtggag ccttg tcacc aacctctaac tgcagaactg qqatgtqqaq ctggaagtgc 961 ctcctcttct gggctgtgct ggtcacagcc acactctgca ccgc aggcc gtccccgacc 1021 ttgcctgaac aagcccagcc ctggggagcc cctgtggaag tggagtcctt cctggtccac 1081 cccqgtgacc tge geaget tegctgtegg ctgcqqgacg atgtgcagag cabcaactgg 1141 ctgcggqacg ggqtgcagc ggeggaaage aaccgcaccc gcatcacagg qqaggaggtg 1201 gagqtgcagg aetcc t c cgcagac cc ggcctctatg ettgegtaac cagcagcccc 1261 tcgggcagtg acaccaccta cttctccgtc aatgtttcag atgctctccc ctcctcggag 1321 gatgatgatg a gatgatcja ctcctcttca cjaggagaa g aaac gataa caccaaacca 1381 aaccgtatgc ceg agetec atattggaca tccccagaaa aqatggaaaa gaaattgcab 1441 gcagtgccgg c qeeaagac agtgaagttc aaatgecett ccag tgggac cccaaacccc 1501 acactgcgct ggttgaaaaa tggcaaagaa ttcaaacctg accacagaat tggaggctac 1561 aaggtccgtt atgccacctg gagcatcata atggactctg tggtgccctc tgacaagggc 1621 aactacacct gcattgtgcja gaatcjagtac ggcagcatca accacacata ccagctggat 1681 gtcqtggagc ggtcccctca ccggcccatc ctgcaagcag gqttgcccqc caacaaaaca 1741 gtggccctgg gtageaacg t ggag ttcatg tgtaaggtqt acag tgaccc qcagccgcac 1801 atccagtggc taaagcacat cgaggtgaat qggagcaaga ttggcccaga caacctqcct 1861 tatgtccacja tcttcjaagac tcjctggacjtt aataccaccg acaaagacjat ggaggtgctt 1921 cacttaagaa atgtctcctt tgaggacgea cjcjggagtata cgtcjcttggc gggtaactct 1981 atcggactct ccca cactc tgcatggttg accgttctgg aagccctgqa agagaggecg 2041 gcagtgatga cctcgcccct gtacctggag atcatcatct attgeacagg qqccttcctc 2101 atetcctgca tggtggggtc ggtcatcgtc tacaagatga agagtgg ac caagaagagt 2161 gacttccaca gecagatgge tcjtgcacaaq ctgcjccaaga cjcatccctct gcgcagacag 2221 gtgtctgctg actccagtcjc atccatgaac tctggggttc ttctggttcg gccatcacgg 2281 ctctcctcca gtgggact.ee catgetagea ggggtctctg aqtatgagct tcccqaagac 2341 cctcgctggg agctgcctcg ggacaqaetg g tcttaggca aacccctggg aqagggctgc 2401 tttgggcagg tggtg tggc agaggctatc gggc ggaca aggacaaacc caaccgtgtg 2461 accaaagtcjc; ctgtgaagat cjttgaag cg gaccjcaacag agaaagactt gtcagacctcj 2521 atctcagaaa gqagatgat gaagatgatc gggaagcata agaatatca caacc gctg 2581 gggqectgea cgcaggatgg cccttg at gtcatcgtgg aqtatgeetc caagggcaac 2641 ctgcggqagt acctgcaggc cggagg cc ccagggc qg aatactgeta caaccccagc 2701 cacaacccag aggagcagct ctcctccaag gacc ggtgt cctgcgccta ccaggtggcc 2761 ccjaqgcatcjcj agtatctggc ctccaagaag tgcatacacc gagacctggc agecaggaat 2821 gt.cctggtga caqaggacaa tgtgatgaag atagcagact ttggcctcgc aegggacatt 2881 caccacatcg actactataa aaagacaacc aacggccgac tqcctg gaa gtggatggca 2941 cccgaggcat tatttgaccg gatctacacc caccagacjtg atgtgtggtc tttcggggtg 3001 ctcctgtggg aga cttcac tctgggcggc tecccatacc ccggtg gcc tgtggaggaa 3061 cttt caagc tgctgaagga ggg caccgc atggacaagc ccagtaactg caccaacgag 3121 ctgtacatga tgatgcggga ctgctggcat gcag gccct cacagagacc caccttcaag 3181 cagctggtgg aagacctgga ccgcatcgtg gccttgaccfc ccaaccagga gtacctggac 3241 ctgtccatgc ccctggacca gtactccccc agctttcccg acacccggag ctctacgtgc 3301 tcctcagggg agga tccgt cttctc cat gagccgctgc ccgaggagcc ctgcctgccc 3361 cgacacccag cccagcttgc caatggcgga ctcaaacgcc gctgactgcc acccacacgc 3421 cctccccaga ctccaccgtc agctgtaacc ctcacccaca gcccctgctg ggcccaccac 3481 ctgtccgtcc ctgtcccctt tcctgctggc aggagcegge tgcctaccag gggccttcct 3541 gtgtggcctg ccttcacccc actcagctca cctctccctc cacctcctct ccacctgctg 3601 gtgagaggtg caaagaggca gatctt gct gccagccact tcat ccctc ccagatgt g 3661 gaccaacacc cctccctgcc accaggcact gcctggaggg cagggagtgg gagecaatga 3721 acaggcatgc aagtgagagc ttcctgagct ttctcctgtc ggtttg tct gttttgeett 3781 cacccataag cccctcgcac tctggtggca ggtgccttgt cctcagggct acagcagtag 3841 ggaggtcagt gcttcgtgcc tcgattgaag cjtgacctctg ccccagatag gtggtgccag 3901 ggctta a attccgatac agtttgctt tgc gaccaa atgc ggta ccagagga g 3961 gtgaggcgaa ggccaggtfcg ggggcagtgt tgtggccctg gggcccagcc ccaaactggg 4021 ggctctgtat atagctatga agaaaacaca aagtgtataa atctgag at atatttacat 4081 gtctttttaa aaggcjtcgtt accagagatt tacccatcgg cjtaagatcjet cctcjgtggct 4141 gggaggcatc a.cjttgcta.ta tattaaaaac aaaaaagaaa aaaa agaaa atgtttttaa 4201 aaaggtcata tat ttttgc acttt gct gt ttattt tttaaa tat gttctaaacc 4261 tattttcagt ttaggtccct caataaaaat tgetgetget tcatttatct atgggctgta 4321 tgaaaagggt gggaatgtcc actggaaaga agggacaccc acgggccctg gggctaggtc 4381 tgtcccgagg gcacccjcatg ctcccggccjc aggttccttg taacctcttc ttcctaggtc 4441 ctgcacccag acctcacgac gcacctcctg cctctccget gc ttggaa agtcagaaaa 4501 agaagatg c tgc cgagg gcaggaaccc ca ccatgca gtagaggege tgggcagaga 4561 tcaagg cc agcagccatc gaccatggat ggtttcctcc aaggaaaccg gtggggfctgg 4621 gctggggagg gggcacctac ctaggaatag ecaeggggta gagctacagt gattaagagg 4681 aaagcaaggg cgcggttgct cacgcctcjta atcccagcac tttgggacac cgaggtgggc 4741 agatcacttc acjgtcaggag tttgagacca gcctggccaa cttagtgaaa ccccatctct 4801 actaaaaatg caaaaattat ccaggcatgg tggcacacgc ctgtaa ccc agctccacag 4861 gaggc;;gagg cagaatccct tgaagctggg aggeggaggt tgcagtgagc egagattgeg 4921 ccattgcact ccagcctggg caacagagaa aacaaaaagg aaaacaaatg atgaaggtct 4981 gcagaaactg aaacccagac atgtgtctgc cccctctatg tgggcatggt tttgccagtg 5041 cttc aagtg caggagaaca tgtcacctga ggctagtttt gca tcagg ccctggcttc 5101 gtttcttgtt ggtafcgcctc cccagatcgt cctfccctgta tccatgtgac cagactgtat 5161 ttgttgggac tgtcgcagat cttggcttct tacagttctt cctg tccaaa ctccatcctg 5221 tccctcagga acggggggaa aattctccga atgtttttgg ttttttggct gcttggaatt 5281 tacttctgcc acctcjctggt catcactgtc ctcactaagt ggattctggc tcccccgta.c 5341 ctcatggctc aaactaccac tcc cagtcg ctatat aaa gc tatatt tgctggatta 5401 ctgctaaata caaaagaaag fctcaatafcgt tttcatttct gtagggaaaa tgggattget 5 61 gctttaaatt tctgagctag ggatfcttttg geagctgeag tgtfcggcgac tattgtaaaa 5521 ttctctttgt ttctctctgt aaatagcacc tgctaacatt acaatttgta tttatgttta 5581 aagaaggcat catttggtga acagaactag gaaatgaatt tttagctctt aaaagcattt 5641 gctt gagac cgcacaggag tgtctttcct gtaaaacag tga gataa ttctgccttg 5701 gccctacctt gaagcaatgt fcgtgtgaagg gatgaagaat ctaaaag bet tcataagt.ee 5761 ttgggagagg tgctagaaaa atataaggca ctatcataat tacagtgatg tecttgetgt 5821 tactactcaa atcacccaca aatttcccca aagactgege tagctgtcaa ataaaagaca 5881 gtcjaaattcja cctga
[00134] SEQ ID NO: 185 is the FGFRl Amino Acid Sequence for isoform 1, having Geneba k Accession No. NP_075598 (822 aa):
1 MWSWKCLLFW AVLV ATLC ARPSFTLPEQ AQPWGAPVEV ESFLVHPGDL LQLRCRLRDD 61 VQSINvvLRDG VQLAES RTR ITGEEVSVQD SVPADSGLYA CV SSPSGSD TTYFSV VSD 121 ALPSSEDDDD DDDSSSSEKE TDNTKP R P VAPYWTSPEK EKKLHAVPA AKTVKFKCPS 181 SGTPNP LRW LKNGKEFKPD HRIGGYKVRY A BSIIMDSV VPSDKG YTC IVENEY6SIN 241 HTYQLDVYER SPHRP1LQAG LPAKKTVALG SilVEFMCKVY SDPQPHXQWL KHIEVNGSKI 301 GPDNLPYVQI LKTAGVNTTD KEMEVLKLRN VSFEDAGEYT CLAGNSIGLS HHSAWLTVLE
361 ALEERPAVMT SPLYLEIIIY CTGAFLXSCM VGSVIVYKMK SGTKKSDF.HS QMAV.HKL.AKS
421 IPLRRQVTVS ADSSAS NSG VLLVRPSRLS SSGTPMLAGV SEYELPEDPR WELPRDRLVL
481 GKPLGEGCFG QWLAEAIGL DKDKPNRVTK VAVKMLKSDA TEKDLSDLIS EMEMMKMIGK
541 HK IINLLGA CTQDGPLYVI VEYASKG LR EYLQARRPPG LEYCYNPSHK PSEQLSSKDL
601 VSCAYQv'ARG MEYLASKKC1 HRDLAARNVL VTED VMKIA DFGLARDIHH IDYYKKTTKG
661 RLPVKWMAPE ALFDR1YTHQ SDVBSFGVLL WETFTLGGSP YPGVPVEELF KLLKEGHRMD
721 KPSNCTNELY MMMP.DCWHAV PSQRPTFKQL VEDLDRIVAL TSNQEYLDLS MPLDQYSPSF
781 PDTRSSTCSS GEDSVFSHEP LPEEPCLPRH PAQLANGGLK RR
[00135] SEQ ID NO: 186 is the FGFR1 Nucleotide Sequence for isofomi 1, having
Genebank Accession No. NM_0231 10 (5917 bp): i aga gcaggg gcgcaaacqc caaaqgagac caggctgtag gaaqagaagg gcagagcgcc
61 ggacage eg gcccgctccc cgtcctttgq ggccgcggct ggggaactac aaggcccagc 121 aggcagctgc agggggegga ggeggaggag ggaccagcgc gggtgggagt gagagagega 181 gccctcgcgc cccgccggcg cata.cjcgctc ggagegctet tgccjcjccaca ggcgcggcgt 241 cctcggcgcjc gggeggcage tagegggage cggcjacgccg gtgcagccgc agegegegga 301 ggaacccggg gtgc ggga getgggegge cacgtccgga egggaccgag acccctcqta 361 gcgcattgcg gcgacctcgc cttccccggc cgcgagcgcg ccgctgcttg aaaagccgcg 421 gaacccaagg acttttctcc ggtccgagct cggggcgccc cgcagggcgc acggtacccg 481 tgctgcagtc gggcacgccg cggcgccggg gcctccgcag ggcgatggag cccggtctgc 541 aaqqaaagtg aggcgccgcc getgegt et ggagqagggg qgcacaaqgt ctggagaccc 601 cgggtggcgg acgggagccc ccccccgee ccgcctccgg ggcaccagct ccggctccat 661 tg ttcccgcc egggctqgag gcqccgagca cga c cg cegggag teg agcgccggcc 721 geggagctet tgcgaccccg ccaggacccg aacagagccc gggggcggcg ggccggagcc 781 ggggacgegg gcacacgccc gctccjcacaa gccacggcgg actctcccga ggeggaaect 841 ccacgccgaq cgaggqtcag tttgaaaaqg aggatcgagc tcactgtgga gtatccatgg 901 agatgtqgag ccttgtcacc aacc ctaac gcagaactg ggafcgtggag ctggaag tgc 961 ctcctcttct gggc gtgct qgtcacaqcc acactctgca ccgctaggcc gtccccgacc 1021 ttgectgaac aagcccagcc ctggggagcc cctgtggaag tggagtcctt cctggtccac 1081 cccggtgacc tcjctgcagct tegctgtegg ctgcgggacg atgtgcagag catcaactgg 1141 ctqcgggacq gggtgcagct gqcggaaaqc aaccqcaccc qcat acagg ggaqgaggtg 1201 gaggtgeagg actccgtc!cc cgcaciac cc qgcctctatg ettqegtaac cagcacicccc 1261 tegggcagtg acaccaccta cttctccgtc aatgtttcag atgctctccc ctcctcggag 1321 gatgatgatg atgatgatga ctcctcttca gaggagaaag aaacagataa caccaaacca 1381 aacegtatge ccgtagctcc atat ggaca tccccagaaa agatggaaaa gaaattcjeat 1441 gcagtgccgq ctgccaagac aqtgaagttc aaatqecett ccagtgggac cccaaacccc 1501 acactqcgct ggttgaaaaa tggcaaagaa bcaaacctg accacagaat tggaggctac 1561 aaggtccgtt atgccacctg gagcatcata atggactctg tggtgccctc tgacaagggc 162.1 aactacacct gcat cjtgga cjaatgagtac ggcagcatca accacacata ccacjctggat 1681 gtcgtggagc ggtcccctca ccggcccatc ctgcaagcag ggttgcccgc caacaaaaca 1741 g qgccc gq gtagcaacgt ggagttcatg tgtaaggtgt acagtgaccc gcaqccgcac 1801 atccagtggc taaagcacat cgaggtgaat gggagcaaga ttggcccaga caacctgcct 1861 tatgtccaga tcttgaagac tgctggag t aataccaccg acaaagagat ggaggtgctt 1921 cacttaagaa atgtctcctt tcjaggaccjca ggggagtata cgtgcttggc gggtaactct 1981 ateggactet cccatcactc tgcatggttg accgttctgg aagccctgga agagaggecg 2041 gcagtgatga cctcgcccct gtacctggag atcatcatct attgeacagg ggccttcctc 2101 atctcctgca tggtggggfcc ggtcatcgtc tacaagatga agag tggtac caagaagagt 2161 gacttccaca gccagatggc tgtgcacaag ctggccaaga gcatccctct gcgcagacag 2221 gtaacagtgt ctgctgactc cagtgcatcc atgaactctg gggttcttct gg tcggcca 2281 tcacggctct cctccagtgg gactcccatg ctagcagggg tctctgagta tgagct ccc 2341 gaagaccc c gctggqagct gcctcgggac agactggtct tagg aaacc cctqggagag 2401 ggctgctttg ggcaggtggt gttggcagag qctatcgqgc tggacaagga caaacccaac 2461 cg gtgacca aagtggctgt gaagatgt g aagtcggacg caacagagaa agacttgtca 2521 gacctgatct cagaaatgga cjatgatgaag atgatcggga agcataagaa tatcatcaac 2581 ctgctggggg cctgcacgca ggatggtccc ttgtatgtca tcgtqgagta. tgc tccaag 2641 ggcaacc gc gggagtacct gcaggcccgg aggcccccag qgctggaata ctgctacaac 2701 cccagccaca acccagagga gcagctct.ee tccaaggacc tggtgtcctg cgcctaccag 2761 gtggcccgag gcatggagta tctggcctcc aagaagtgca tacaccgaga cctggcagcc 2821 aggaatgtcc tggtgacaga ggacaatgt.g atgaagatag agactttgg cctcgcacgg 2381 gacattcacc acatcgacta c ataaaaag acaaccaacg gccgac gcc tgtgaagtgg 2941 atggcacccg aggcattatt tgaceggate tacacccacc agag gatgt gtggtctttc 3001 ggggtgctcc tcjtgggagat cttcactctg ggcggctccc cataccccgg tgtgcctgtg 3061 gaggaac t teaagctget gaaggagggt caccgcatgg acaagcccag taactgcacc 3121 aacgagctgt acatgatga gcgggactgc tggcatgcag tgccctcaca gagacccacc 3181 ttcaagcagc tggtggaaga cctggaccgc atcg ggcct tgacctccaa ccaggagtac 3241 ctggacctgt ccatgcccc ggaccagtac tcccccagct ttcccgacac ccggagctct 3301 acgtgctcct caggggagga ttccgtcttc tctcatgagc cgctgcccga ggagccctgc 3361 c gccccgac acccagccca gettgecaat ggeggactea aacgccgctg actgccaccc 3421 acacc!ccetc cccagactcc accg cagct g taaccc ca cccacagccc ctgctgggcc 3481 caccacctgt ccgtccctgt cccctttcct gctggcagga gccggctgcc taccaggggc 3541 cttcctgtgt ggcctgcctt caccccactc agctcacctc tccctccacc tcctctccac 3601 ctgctggtga gaggtgcaaa gaggcagatc tttgetgeca gccacttcat cccctcccag 3661 atgttggacc aacacccctc cctgccacca ggcactgcct ggagggcagg gagtgggagc 3721 caatgaacag geatgeaag t gagagcttcc tgagctttct cctg teggtt tggtctg tt 3781 tgccttcacc cataagcccc tcgcactctg gtggcaggtg ccttgtcctc agggctacag 3841 cagtagggag gtcagtgctt ccjtgcctccja ttgaaggtga cctctgcccc agataggtgg 3901 tgccacjtggc ttattaattc cgatactagt ttgetttget gaecaaatge ctggtaccag 3961 aggatgg ga ggegaaggee aggttggggg cagtgttgtg gccct.ggggc ccagccc aa 4021 actgggggct. c gtatatag ctatgaagaa aacacaaagt gtataaatct gagtata at 4081 ttacatgtct ttttaaaagg gtcgttacca gaga ttacc catcggg aa gatgetcctg 4141 gtggctggga ggcatcagtt ejetatatatt aaaaacaaaa aaaaaaaaaa agejaaaatgt 4201 ttttaaaaag g catata t ttttgetact tttg tgttt tattttttta aatt.atgttc 4261 taaaccta t ttcagtttag gtccctcaat aaaaattgct getgettcat ttatctatgg 4321 gctgtatgaa aagggtggga atgtccactg gaaagaaggg acacccacgc; gccctggggc 4381 taggtctgtc ccgagggcac cgcatgctcc eggegcaggt tccttgtaac ctcttcttcc 4441 tacjgtcctcjc acccacjacct cacgacgcac ctcctgcctc tccgctcjctt ttcjcjaaagtc 4501 agaaaaagaa gatgtctget tcgagggcag gaaccccatc catgeagtag aggcgctggg 4561 cagagagtca aggcccagca gccatcgacc at.ggatggtt tcct caagg aaaccggt.gg 4621 ggttgggctg gggagggggc acctacctag gaatagecac gggg tagagc tacagtgatt 4681 aagaggaaag caagggegeg g tgctcacg cctg aatcc cagcactttg ggacaccgag 4741 gtgggcagat cacttcaggt caggagtttg agaccagcct cjgccaactta gtcjaaacccc 4801 atctctacta aaaatgcaaa aattatccag gcatggtggc acacgcct.gt aatcccagct 4361 ccacaggagg ctgaggcaga atcccttgaa gctgggaggc ggaggt gca gtgagecgag
4921 attgegecat tgcactccag cctgggcaac agagaaaaca aaaaggaaaa caaatga ga 4981 aggtctgeag aaactgaaac ccagacatgt gtctgccccc tctatgtggg catggttttg 5041 ccagtgcttc taagtcjcagg acjaacatcjtc acctcjaggct agttttcjcat tcacjgtccct 5101 gg ttcgttt cttgttggta tgcctcccca gategtcett cctgtat ca. tgtgaccaga 5161 ctgtatttg t tgggactgtc gcagatcttg gct cttaca gttcttcctg tccaaact.ee 5221 atcctgtccc teaggaaegg ggggaaaatt ctccgaatgt ttttggtttt ttggctgett 5281 ggaatttact tctgccacct gctggtcatc actg cctca ctaagtggat tctggctccc 5341 cccjtacctca tggctcaaac taccactcct cagtegctat attaaacjett atattttget 5401 ggattactgc taaatacaaa agaaagttca atatgttttc atttctgtag ggaaaatggg 5461 attgetgett taaa tctg agctagggat tttfctggcag ctgcag gtt ggegactat 5521 gtaaaattct. ctttgtttct ctctgtaaat agcacctgct aaca tacaa tttgta ta 5581 tg ttaaaga aggcatcatt tggtgaacag aactaggaaa tgaattttta actcttaaaa
5641 geatttgett tgagaccgea caggagtcjtc tttccttgta. aaacagtcjat ga aatttct 5701 gccttggccc taccttgaag caatgttgt.g tgaagggatg aagaatct.aa aagtcttcat 5761 aagtccttgg gagaggtget agaaaaa at aaggcactat cataat aca gtgatgtcc 5821 tgctgttact actcaaatca cccacaaatt tccccaaaga ctgcgctagc tgtcaaataa 5881 aagacagtga aattgacctg aaaaaaaaaa aaaaaaa
[0(5136] The Genbank ID for the TACCi gene is 6867. Three isoforms are listed for TACC1 , e.g., having Genebank Accession Nos. NP_006274 (corresponding nucleotide sequence NM_001 174063); NP_001167535 (corresponding nucleotide sequence NM 001 174064); NP 001 167536 (corresponding nucleotide sequence NM 001 174065).
100137] SEQ TD NO: 148 is the TACC1 Amino Acid Sequence for isoform 1 , having Genebank Accession No. NP 006274 (805 aa):
1 MAFSPWQILS PVQWAKWTWS AVRGGAAGED EAGGPEGDPE EEDSQAETKS LSFSSDSSGN
61 FETPEAE PI RSPFKESCDP SLGLAGPGA.K SQESQSADEQ LVAEWBKCS SKTCSKPSE
121 EVPQQA DSH SVKNFREEPE HDFSKISIVR PFSIETKDST DISAVLGTKA A.HGCVTAVSG
181 KALPSSPPDA LQDEAMTEGS MGVTLEASAE ADLKAGNSCP ELVPSRR3KL RKPKPVPLRK
241 KAIGGEFSDT KAAVEGTPLP KASYHFSPEE LDE TSPLLG DARFQKSPPD LKETPGTLSS
301 DTNDSGv'ELG EESRSSPLKL EFDFTSDTGN 1EARKALPRK LGRKLGSTLT PKIQKDG1SK
361 SAGLEQPTDP VARDGPLSQT SSKPDPSQWE SPSFNPFGSH Sv'LQNSPPLS SEGSYKFDPD
421 KFDESMDPFK PTT LTSSDF CSPTGNRVNE ILESPKKAKS RLTTSGCKVK KHETQSLALD
481 AC3RDEGAVI SQISDISKRD GHATDEEKLA STSCGQKSAG AEVKGEPEED LEYFECSNVP
541 VSTIKHAFSS SEAGIEKETC QK EEDGST V LGLLESSAE APVSVSCGGE SPLDGICLSE
601 SDKTAVLTLI RESI1TKE1E ANEWKKKYEE TR.QEVLEMRK IVAEYEKTIA QMIEDEQRTS
661 MTSQKSFQQL TMEKEQALAD LNSVERSLSD LFRRYEKLKG VLEGFKKNEE ALKKCAQDYL
721 ARVKQEEQRY QALK HAEEK LDKA EEIAQ VR K.AKAESA ALHAGLRKEQ MKVESLERAL
781 QQKNQEIEEL TKICDELIAK LGKTD
{00138] SEQ ID NO: 149 is the TACCl Nucleotide Sequence for isoform 1, having Genebank Accession No. NM 006283 (7802 bp): i acjctgatgcg cgccccgccg gccgggagqc gggagtccgc gagccgggag egggagcage
61 agagg ctag cagccgggcg ccgcgggccg ggggcc gag gaggccacag qacgggcgt.c 121 ttcccggcta gtggagcccg gcgcggggcc cgctgcggcc gcaccgbgag gggaqgaggc 181 cgaggaggac gcagcgccgg ctgccggcgg gaggaagcgc tccaccaggg cccccgacgg 241 cactcgttta accacatccg cgcctctgct ggaaacgctt gctggcgcct gtcaccggtt 301 ccctccattt tgaaagggaa aaaggc ctc cccacccatt cccctgcccc taggagctgg 361 agccggagga gccgcgc ca tggcgttcag cccgtggcag atectgt.eec ccgtgcagtg 421 ggcgaaatgg acgtggtctg cggtacgcgg cggggccgcc gqcgaggacg aggctggegg 481 gcccgagggc gaccccgagg aggaggattc gcaagccgag accaaatcct tgagtttcag 541 ctcggattct gaaggtaatt ttgagactcc tcjaagctcjaa accccgatcc gatcaccttt 601 caaggagtcc tgtgatccat cactcggatt ggcacjgacct ggggccaaaa gecaagaate 661 acaagaagct gatgaacagc ttgtagcaga agtggttgaa aaafcgttcat ctaagacbtg 721 ttctaaacc tcagaaaatg aagtgccaca gcaggccatt gactctcact cag bcaagaa 781 tttcagagaa gaacctgaac atgattttag caaaatttcc ategtgagge cattttcaat 841 agaaacgaag gattccaccjc; atatctcggc acjtcctccjga acaaaagcag ctcatgcjctg 901 gtaactgca gtc caggca aqgctc gcc ttccaqcccg ccagacgccc tccaqgacga 961 ggcgatgaca gaaggcagca tggggqtcac cctcgaggcc tccgcagaag ctgatctaaa 1021 agctggcaac tcctg tccag agcttgtgcc cagcaqaaga aqcaagcbqa gaaaqcccaa 1081 gcctgtcccc ctgaggaaga aagcaattgg aggagagttc tcagacacca aegctgetgt 1141 ggaggcjcaca cctctcccca aggcatccta tcacttcagt cctgaagagt tggatgagaa 1201 cacaagtcct ttgc aggag atqccaggtt ccagaagtc ccccctgacc ttaaagaaac 1261 tcccggcact cfccagtagfcg acaccaacga c caggggtt gagcbggggg aqgagtcgag 1321 gagctcacc ctcaagcttg agtttgafctt cacagaagat acaggaaaca tagaqgecag 1381 gaaagccctt ccaaggaagc ttggcaggaa actgggtagc acac gactc ccaagataca 1441 aaaagatggc atcagtaacjt cagcaggttt agaacagcct acagacccag tggcacgaga 1501 cgggcctctc tcccaaacat cttccaagcc aga cctag cagtgggaaa gccccagctt 1561 caaccccttt gggagccac ctgtfcctgca gaactcccca cccc bctctt ctgagggctc 1621 ctaccacttt gacccagata actttgacga atccatggat ccctttaaac caactacgac 1681 cttaacaacjc agtgactttt cjttctcccac tggtaatcac cjttaatcjaaa tcttagaatc 1741 acccaagaag gcaaagtccjc gtttaataac cjagtggctgt aagcjtgaaga agcatgaaac 1301 cagtctc c gccctggatg catgttctcq ggabgaaggg gcagtgatct. cccaqatttc 1861 agacatttct aatagggatg gccatqctac bgatgaggag aaacbggcat ccaegtcatg 1921 tggtcagaaa tcagctggtg ccgaggtgaa aggtgagcca gaggaagacc tggagtactt 1981 tgaatcjttcc aatgttcctg tgtctaccat aaatcatcjcg ttttcatcct cagaagcagg 2041 catagagaag gagacgtgcc agaaga gga agaagacggg ccac gbgc t tgggctgct 2101 ggagtcctct gcagagaagg cccctgtgtc ggtgtcc gt ggagg gaga gccccctgga 2161 tgggatctgc ctcagcgaat cagacaagac agccgtgctc accttaataa gagaagagat 2221 aattactaaa csacrattjaacr caa gaatg gaagaagaaa cccggcaaga 2281 agttttggag atgaggaaaa ttgtagctga atatgaaaag actattgctc aaatgattga 2341 agatgaacaa aggacaagta tgacctctca gaagagcttc cagcaactga ccatggagaa 2401 ggaacaggcc ctggctgacc ttaactctgt ggaaaggtcc ctttctgatc tcttcaggag 2461 atatgagaac ctgaaaggtg ttctggaagg gttcaagaag aatgaagaag ccttgaagaa 2521 atgtgctcag gattacttag ccagagttaa acaagaggag cagcgatacc aggccctgaa 2581 aatccacgca gaagagaaac tggacaaagc caatgaagag attgctcagg ttcgaacaaa 2641 agcaaaggct gagagtgcag ctctccatgc tggactccgc aaagagcaga tgaaggtgga 2701 gtccctggaa agggccctgc agcagaagaa ccaagaaatt gaagaactga caaaaatctg 2761 tgatgagctg attgcaaagc tgggaaagac tgactgagac actcccccbg ttagctcaac 2821 agatctgcat ttggctgctt ctcttgtgac cacaattabc ttgccttatc caggaataat 2881 tgcccctttg caqagaaaaa aaaaaactta aaaaaagcac atgcctactg ctgcctcjtcc 2941 cgctttgctg ccaatgcaac agccctggaa gaaaccctag agggt gcat ag ctagaaa 3001 ggagtgtgac cbgacagtgc tggagcctcc bagtttcccc ctabgaaggt bcccttaggc 3061 tgcbgagttb gggttbgtga tbbatcttba gtttgbttta aagtcatcbt tacbbtccca 312.1 a.atgtgttaa atttcjtaact cctctttggg gtcttctcca. ccacctcjtct gatttttttg 3181 tgatctgttt a.atcttttaa ttttttagta tcagtggttt tatttaagga gacagtttgg 3241 cctattg ta cttccaattt abaatcaaga aggggctctg gatccccttt taaattacac 3301 acactctcac acacatacat gtatg bttat agatgctgct gctcbtttcc c gaagcata 3361 gtcaagtaag aactgc beta cagaaggaca tattbccttg gatgtgagac cctabtttga 342.1 aatagagtcc tgactcagaa caccaactta agaatttggg ggattaaaga tgtcjaagacc 3481 acag cttgg gttttcatat ctggagaaga cbatttgcca tgacgttttg btgcccbggt. 3541 a ttggacac tcc cagctt baatgggbgt ggccccttta gggttag cc tcagactaab 3601 gatagbgtct gcbttctgca tgaacggcaa batgggacbc cctccaagct agggtt ggc 3661 aag bctgccc tagagbcatt tactctccbc tgccbccatt bgttaataca gaabcaacat 3721 t agtcttca ttatcttttt tttttttttt gagacagagt ttcgatctat tttaagtatcj 3781 tgaagaaaat ctacttgtaa aaggctcaga tcttaattaa aagcjtaattg tagcacatta 3841 ccaatta aa ggtgaagaaa gtttt c ccaagtgtga bgcat g tc ttcagatg 3901 gaaaagaaag caaaaaatac cttc aactt aagacagaat tttbaacaaa a gagcagta 3961 aaagtcacab gaaccactcc aaaaatcagt gcatbbtgca batttttaaa caaagacagc 4021 ttgttgaata ctgagaagag gagtgcaagg agaaggtctg tactaacaaa gccaaattcc 4081 tcaagctctt actggac ca gttcagagtg gbgggcca t aaccccaaca bggaa tt 4141 ccabataaab ctcaabgaat bccctttcat ttgaa aggc aaacccaaat ccabgcaagb 4201 gttttaaagc acbgtcctgb cttaabctta catgctgaaa gtc bcatgg bgatatgcac 4261 tabattcag atacgbatgt tbbcctacbb ctctbgtaaa actgttgcat gatccaactt 432.1 cacjcaatgaa ttgtcjcctag tcjgagaacct ctatagatct taaaaaatga attattcttt 4381 agcag gtat tactcacatg ggtgcaatct agccccag ggaggtcaat aatgtcbbtt 4441 aaagccagaa gtcacatttt accaata gc att abcata a tggtgc t aggcbgtata 4501 ttcaagcctg t gtcttaac atttbgtata aaaaagaaca acagaaatta bctgtcabtt 4561 gagaagtggc ttgacaatca tbbgagctbb gaaagcagtc actgtgg gt aatabgaatg 462.1 ctgtcctagt ggtcatagta ccaagggcac gtgtctcccc ttggta.taac tgatttcctt 4681 tttagtcctc actgctaaa taag aatt gcat g cagaaagaaa cattga gc 4741 taaatcttbb tgctgctgtg bbtggtgbt; ttcabgttta cbtgtt bat atbgatctgb 4801 tttaagtatg agaggcttab agtc!ccc cc abtgtaaabc catagtcatc tttttaagct 4861 tabbgtgttb aagaaagtag c atgtgt a aacagaggtg abggcagccc ttccctagca 4921 cactggtgcja agagacccct taagaacctg accccagtga atgaagctga tgcacaggga 4981 gcaccaaagg accttcg ba agtga aatt gbcctggcct ctcagccatg accgt a ga 5041 ggaaatatcc cccafcbcgaa cb aacagat; gcc cctctc caaagagaat taaaatcgta 5101 gcttgtacag atcaagagaa tatac gggc agaatgaagt atgtbtgttt abttttcbtt 5161 aaaaataaag gatttbggaa c ctggagag taagaatata g atagagtt tgccbcaaca 5221 catgtgaggg ccaaataacc bgctagcbag gcagbaataa actctg ac agaagagaaa 5281 aagggccggg cacagtggc tat cctgta atcccaacac tg ggaaggc cgaggcagga 5341 ggabcactbg agtccaggag bbgaaacct; acc aggcaa catggtgaaa ccbbgtctcb 5401 accaaaataa aaattagctg ggcatggtgg cacgtgcc g tggtcccagc bacttgggag 5461 gctgaggtgg gagcc ggga gg bcaaggcb gcag gagcc abgatcabgc cac gcactc 5521 catcctgggt gacacjcaaga tcttgtctca aaaaaaaaaa aaaaaaaaaa aaaaccagga 5581 gtgaaaaagg aaagtagaag gcagc gctg gcctagatgt tgg tggga atattaggtg 5641 atcctgttga gattctggat ccagagcaat ttctttagct tttgactttg ccaaagtcjta 5701 gatagcc t atccaqcagt attttaagtq gggaatgcaa cgtgaggcca actgaacaat 5761 tccccccgt.g gctgcccaga tagtcacagt caaggttgqa gag ctcctt ccagccagtg 5321 acctacccaa acctt tgtt c qtaaaact gctc qgaaa taccgggaag cccaqttttc 5881 tcacgtggtt tctagcttct tcagactcag cccaaattag gaagtgcaga agcacatgat 5941 ggtgaaaaac ctaggatttg gcagccttcc acjaatggtat ggaatctgag ggaagattta 6001 gtttcg t tggaggatag ctcaagttga att tctttc cagccagtta ccctttcaac 6061 ctacccatac tttgtacaac tcttacacaa a acttagat atttattaga tagccctgaa 6121 ttcactctaa ttataaacag ggagtgtaaa ctgcccccag atgttcctgg gctgggtaaa 6181 agcagctgqa gtgaagcact cattttccat aaaggtaaca aagggcagct cagtggttac
6241 tcaagctcaa a.agggttttt ttaacjagcaa cjcattggtta agtctgtgta tactgagttg 6301 gaagtga tt cagcacattc tttttagtq gag gaaagt tctgaagccc ccttttaact
6361 tcctcfctggt ttttcattat aattgqtagc catctcatqa actg tctctg actgttg bet 6421 ctttgtggtc atgtgattgt gagcttgctt tctgacttgc atttctgact ttatcctgtt 6481 gttaggaaga tagaaactag qttttgaaag attacatgat tcaagcgagg gattttaaag 6541 taaagatgta tttattctga agaatctaaa acjataacaga ttatttgctt atgaaacjaac 6601 aatatag ct gggaatccca gaatgtcaaq ccaaaqgtct aagaag cat ctccttcaaa 6661 tactttaata aaqaagtatb tcgaggagat a ctgtccaa aaaggtttga ctggcctcca 6721 gattccagtt atttttaaaa agcaacttac cactaaatcc ttgagtctcc atagagtaac 6781 acjtaaagaaa ctgatgtaac agactctcct ctcaaaggat ctcctctcjga agacjactatc 6841 agcggcagca t ctccagcjc; aagacccatc ccctagtcjcc agacjcttgca tcctggagac 6901 aaagat gc act tttgt agttttt gt ccaaatgcaa tcccat tct gtgectctta 6961 gcatgcagtt agatttggac aaacaagatt cctaaggaat gact tatta actataa at 7021 ggttacagct attatataaa tatatattct ggttatagtt ctaatatgga gatgttgtgt 7081 gcaatgctcjc; cctgtggtgg tctgtgtaat gctttaactt gtatggagga ggccaggctc 7141 agagctgaga tgtggcctga accttccctg atcgatcct ttaatttaga actgtcaaga 7201 tgtcactttc tccccctctg ccttttagtq gta ctgaca tatactcaaa acagtaatt 7261 cctgc! tcaca tcattaactg ctaattctgt a ttataaag aatt tcaga tqgacatgta 7321 caaatttgaa ctcaaaccat ccccagtcca gatacagggc agcgtgtagg tgaccacacc 7381 agagcctcag cctcqqtcct tctcagcccjt cgggatagga tccaggcatt tcttttaaat 7441 ctcagaggta gcagtaaact tttcagtatt cjctgtta.cjca agtcjtgtgtt tgecaataga 7501 acccat at actaatqtgc caagtaaatq ttcattgcac atctgc tcc actgtgttcc 7561 cacgggtgcc atqaagtgtg tgaggagccc c catctgqa ggga gagtg ctgcgttgac 7621 tactgctatc aggattgtgt tgtgtggaat attcatctac ataaatttta tatgeacagt 7681 aatttccctt ttta.tatgtc aagtaactat ttgtaaaagt tatactcaca aattattata 7741 atga tacta atatattttt tccatqtttc attgcctgaa taaaaactgt ttaccactgt 7801 ta
[00139] SEQ ID NO: 150 is the amino acid sequence of the FGFR1-TACC1 fusion protein.
MWS KCLLFWAVLVTATLCT/RP
AVPAAKTVKFKCPSSGTPNPTLRHLKNGKEFKPDHRIGGYKVRYATWSII DSVVPSDKGNYTCIVSNEYGSIKH TYQLDWEi^PHRPIX^AGLPA KTVALGSN FMCKVYSDPQPHXQWLKHIEWGSKIGPDNLPYVQILKTAGV NTTDKE E\/LHLRNVSFEDAGEYTCLAGNS1GLSHF1SABLTVLEALEERPAVM?SPLYLEIIIYCTGAFLISCMV GSVI\/YKMKSGTKKSDFHSQMAVHKLAKSIPLRRQy?VSADSSASM SGVLLVRPSRLSSSGTP LAGVSEYELP EDPRwELPRDRLVLGKPLGEGCFGQn/LAEAIGLDKDKPN
K IINLLGACTQDGPLYVIVEYASKGNLREYLQARRPPGLEYCY PSHNPEEQLSSKDLVSCAYQVARGMEYLAS
KKCIHRDLAAR VLVTED V KIADFGLARDIHHIDYYKKTTKGRLPVKKMAPEALFDRIYTHQSDVwSFGV^
EIFTLGGSPYPGVPVEELFKLLKEGHP^IDKPS CT ELYM MRDC HAVPSQRPTFKQLVEDLDRIV^
LSSSAS APVSVSCGGESPLDGICLSESDKTAVLTLIREEIITKEIEANEWKKKYEETPJQEVLEMRKIVAEYEKT
lAQMIEDEQRTS TSQKSFQQLTMEKEQALADLNSVERSLSDLFRRYENLKGVLEGFKKKEEALKKCAQDYLARV
KQEEQRY'QAiKIHAEEKLDKANEEIAQVRTFAKAESA^ HAGLRKEQMKVESLERA
AKLGKTD
{00140] SEQ TD NO: 151 is the nucleotide sequence that encodes the FOFR 1 -TACC 1 fusion protein. atgtggagctggaagtgcctcctcttctgggctgtgctggtcacagccacactctgcaccgctaggccgtccccg accttgcctgaacaagcccagccctggggagcccctgtggaagtggagtccttcctggtccaccccggtgacctg ctgcagcttcgctgtcggctgcgggacgatgtgcagagcatcaactggctgcgggacggggtgcagctggcggaa agcaaccgcacccgcatcacaggggaggaggtggaggtgcaggactccgtgcccgcagactccggcctctafcgct tgcg aaccagcagcccc cgggcag bgacaccacctac bctccg caatgt bcagatgctctcccctcctcg gaggatgatgatgatgatgatgactcctcttcagaggagaaagaaacagataacaccaaaccaaaccgtatgccc gtagctccatattggacatccccagaaaagatggaaaagaaattgcatgcagtgccggctgccaagacagtgaag ttcaaatgcccbbccagtggqaccccaaaccccacactgcgctggttgaaaaabgqcaaagaattcaaacctgac cacagaat ggaggc acaagg ccgtta gccacc ggagca cataatggactctg bggtgccctctgacaag ggcaactacacctgcattgtggagaatgagtacggcagcatcaaccacacataccagctggatgtcgtggagcgg tcccctcaccggcccatcctgcaagcagggttgcccgccaacaaaacagtggccctgggtagcaacgtggagttc atgtgtaaggtgtacagtgacccgcagccgcacatccagtggctaaagcacatcgaggtgaatgggagcaagatt ggcccagacaacctgcc 1atgtcc gatc1 gaagactgctggag11aataccaccgacaaagaga gqagg g c tcacttaagaaatgtctcct tgaggacgcaggggagtatacgtgcttggcgggtaactctabcggactcbcc catcactctgcatggttgaccgttctggaagccctggaagagaggccggcagtgatgacctcgcccctgtacctg gagatcatcatctattgcacaggggccttcctcatctcctgcatggtggggtcggtcatcgtctacaagatgaag agtggtaccaagaagagtgacttccacagccagatggctgtgcacaagctggccaagagcatccctctgcgcaga caggbgtctgctgactccagtgcatccatgaactctgggg11cttctgg11cggccatcacgqctctcctccagb gggactcccatgctagcaggggbctctgag batgagcttcccgaagacccbcgctgggagctgcctcgggacaga ctggtcttaggcaaacccctgggagagggctgctttgggcaggtggtgttggcagaggctatcgggctggacaag gacaaacccaaccgtgtgaccaaagtggctgtgaagatgttgaagtcggacgcaacagagaaagacttgtcagac ctgatctcagaaatggagatgatgaagatgatcgggaagcataagaatatcatcaacctgctgggggcctgcacg caggatggtccc11gta gtcatcgbgqagtabgcctccaaqggcaacctgcgggagtacctgcaggcccggagg cccccagggctggaabactgctacaaccccagccacaacccagaggagcagctct.ee ccaaggacctggtg tec tgcgcctaccaggtggcccgaggcatggagtatctggcctccaagaagtgcatacaccgagacctggcagccagg aatgtcctggtgacagaggacaatgtgatgaagatagcagactttggcctcgcacgggacattcaccacatcgac tactataaaaagacaaccaacggccgactgcctgtgaagtggabggcacccgaggcab atttgaccgg ebac acccaccagagtgatgtgbggtctttegggg g:tcctgtgggagatcttcactctgggeggetccccatacccc ggtgtgcctgtggaggaacttttcaagctgctgaaggagggtcaccgcatggacaagcccagtaactgcaccaac gagctgtacatgatgatgcgggactgctggcatgcagtgccctcacagagacccaccttcaagcagctggtggaa gaccbgqaccgcatcgtggccttgacctccaaccagtgggctgctggagtcctctgcagagaaggcccctgtgtc ggtgtcGtgtggaggtgagagccGcctggatgggatctgcctcagcgaatcagacaagacagccgtgctcacGtt aataagagaagagataattactaaagagattgaagcaaatgaatggaagaagaaatacgaagagacccggcaaga agttttggagatgaggaaaattgtagetgaatatgaaaagactattgetcaaatgattgaagatgaaeaaaggae aagtatgacctctcagaagagcttccagcaactgaccatggagaaggaacaggcccggctgacc taacctgt ggaaaggtccctttctgatctcttcaggagatatgagaacctgaaaggtgttctggaagggttcaagaagaatga agaagecttgaagaaatgtgctcaggattacttagccagagttaaacaagaggagcagcgataccaggecctgaa aatccacgcagaagagaaaGtggacaaagccaatgaagagattgctcaggttGgaacaaaagcaaaggGtgagag tgcagcctccatgctggacccgcaaagagcagatgaaggtggagtccctggaaagggcccgcagcagaagaa ccaagaaat.fcgaagaactgacaaaaat.cfcgtgat.gagctgat.fcgcaaagctgggaaagactgac
[00141] The Genbank ID for the FGFR2 gene is 2263. Eight isoforms are listed for
FGFR2, e.g., having Genebank Accession Nos. NP 000132. (corresponding nucleotide sequence M_000141); NP_001 138385 (corresponding nucJeotide sequence
NM_001144913); NP_001 138386 (corresponding nucleotide sequence NM 001 144914); NP 001 138387 (corresponding nucleotide sequence NM 001 144915); NP 001 138388 (corresponding nucleotide sequence NM 001144916); NP 001 138389 (corresponding nucleotide sequence NM__001 144917); NP_G01 138390 (corresponding nucleotide sequence NM_001 14491 8); NP_001138391 (corresponding nucleotide sequence NM_001 144919); NP__075259 (corresponding iiucleoiide sequence NM__ 022970). SEQ ID NO: 152 is the FGFR2 Amino Acid Sequence for isoform 1, having Genebank Accession No. NP 000132 (821 aa):
1 MVSKGRFICL VWTMA LSL ARPSFSLVED TTLEPEEPPT KYQISQPEVY VAAPGEΞLE 61 RCLLKDAAV1 SWTKDGVHLG PNNRTVLIGE YLQIKGATPR DSGLYACTAS RTVDSETWYF 121 MVNVTDAISS GDDEDDTDGA EDFVSE S N KRAPYWTNTE KMEKRLHAVP AANTVKFRCP 181 AGG PMPT R LKNGKEFKQ EHRIGGYKVR NQHWSLIMES WPSDKGNYT CWE EYGSI 241 NHTYHLDWE RSPRRPILQ GLPAMASTVV GGDVEFVCKV YSDAQPHIQW KHVEKNGSK 301 YGPDGLPYLK VLKAAGVNTT DKSIEVLY1R NVTFEDAGEY TCLAG S1GI SFHSAKLTVL 361 PAPGREKEIT ASPDYLEIA1 YCIGVFLIAC MVVTVILCRM KNTTKKPDFS SQPAVHKLTK 421 RIPLRRQVTV SAESSSSMNS TPLVRITTR LSSTADTPML AGVSEYELPE DPK EFPRDK 481 LTLGKPLGEG CFGQWMAEA VGIDKDKPKE AVTVAVKMLK DDATEKDLSD LVSE EMMKM 541 T.GKHKNT.T.NL LGACTQDGPL YVTVEYASKG LREYLRARR PPGMEYSYDI NRVPEEQMTF 601 KDLVSCTYQL ARGMEYLASQ K.C HRDLAAR ' VLVTEK'VM K1ADFGLARD IN 1DYYKKT 661 TNGRLPVKWM APEALFDRVY ?HQSD BSFG VLMWEIFTLG GSPYPGIPVE ELFKLLKEGH 721 RMDKP NCT ELYMMMRDCW HAVPSQRPTF KQLVEDLDRI LTLTTNEEYL DLSQPLEQYS 781 PSYPDTRSSC SSGDDSVFSP DPMPYEPCLP QYPHINGSVK T
SEQ ID NO: 153 is the FGFR2 Nucleotide Sequence for isoform 1, having Genebank Accession No. NMJ3Q014! (4654 bp): i ggcggcqgct ggaggagagc gcggtggaga gecgageggg egggeggegg qtg ggaqcg
61 ggcgagggag cgcgcgcggc cgccacaaag ctcgggcgcc gcggggctgc atgcqqcgta 121 cctggcccgg cgcggcgact gctctccggg ctggcggggg ccggccgcga gccccggggg 181 ccccgaggcc gcagcttgcc tgcgcgctct gagccttcgc aactcgegag caaagtttgg 241 tcjgaggcaac gccaagcctg agtcctttct tcctctcgtt ccccaaatcc gagggcagee 301 cgcgggcgtc atgcccgcgc tcc ccgcag cctgggg ac gcgtgaagcc cqggaggctt 361 ggcgccggcg aagacccaag gaccactctt ctgcgtttgg agttgc ccc cgcaaccccg 421 ggctcgtcgc tttctccatc ccgacccacg eggggegegg ggacaacaca ggtcgeggag 481 gagcgttgcc attcaagtcja ctgcagcagc agcggcacjcg cctcggttcc tgagcccacc 541 gcaggctgaa ggcattgcgc gtagtccatg ccegtagagg aagtgtcjcag atgggattaa 601 cgtccacatg gagatatgga agaggacegg ggattggtac gtaaccatg gtcagctggg 661 gfccgtttcat ctgcc ggtc g tggtcaeca tggcaacctt gtccctggcc cggccctcc 721 tcagtttagt tgaggatacc acattagagc cagaagagee accaaccaaa taccaaatct 781 ctcaaccaga acjtgtacgtcj gctgcgccag cjcjgagtccjct agacjcjtgcgc tgcctgttga 841 aacjatgcccjc cgtgatcagt tggactaagg atgcjggtgca cttgggcjccc aacaatagga 901 cagtgcttat tggggagtac ttgcagataa agggcgccac gectagagae tccggcctct 961 afcgcttgtac tgccagtagg actgtagaca gtgaaacttg gtacttcatg gtgaatgtca 1021 cagatgccat ctcatccgga gatgatgagg atgacaccga tggtgcggaa gattttg ca 1081 gtgaga cag taacaacaac; agagcaccat actggaccaa cacaqaaaag tggaaa gc 1141 ggctcca gc tgtgcctgcg gccaacactg t aagtttcg ctgcccagcc ggggqgaacc 1201 caatgccaac catgcgg gg ctgaaaaa g ggaaggagtt taagcaggag categcattg 1261 gaggctacaa ggtacgaaac cagcactgga gec cattat ggaaagfcgtg gtcccatctg 1321 acaagggaaa ttatacctgt gtagtggaga a gaataegg gtccatcaat cacacgtacc 1381 acctggatgt tcjtggagccja tcgcctcacc ggcccatcct ccaagccgga ctgccggcaa 1441 a gcctccac agtggtcgga ggagaegtag ag tgtctg caaggt tac agtgatgccc 1501 agccccacat ccagtggatc aagcacgtgg aaaagaaegg cag aaatac qqgcccgacg 1561 ggctgcccta cctcaaggtt c caaggccg ccgg gttaa caccacggac aaagagattg 1621 aggttctcta tattcggaat gtaacttttg aggaegctgg ggaatatacg tgcttggcgg 1681 gtaat ctat tcjggatatcc tttcactctg catggttcjac agttctgcca gcgcctcjcjaa 1741 gagaaaagga gat acaget ccccagact acctggagat aqecat tae tgcatagggg 1801 tcttcttaat cgcctgta g gtgg aacag tcatcctgtg ccgaatgaag aacacgacca 1861 agaagccaga cttcagcagc cagccggc g tgeacaaget gaecaaaegt atccccctgc 1921 ggagacaggt aacagtttcg gctgagtcca gctcctccat gaac ccaac accccgc gg 1981 tgaggataac aacacgcctc tettcaaegg cagacacccc catcjctggca ggggtctccg 2041 agtatgaact tccagaggac ccaaaatggg agt tccaag aqataagctg acactgggca 2101 agcccctggg agaaggttgc tttgggcaag fcggtcatggc ggaagcagtg qqaattgaca 2161 aagacaagcc caaggaggcg gtcaccgtgg ccgtgaacjat gttcjaaagat qatgccacag 2221 agaaagacct ttc gatctg gtqtcagaqa tggaqatgat qaagatgatt gggaaacaca 2281 agaa atcat aaatcttc t ggagcctgca cacaggatqg gcctctctat qtcatagttg 2341 ag tatgcctc taaaggcaac ctccgagaat acctccgagc ccggaggcca cccgqgatgg 2401 agtactccta tgacattaac cgtgttcctg aggagcagat gaccttcaag gacttggtgt 2461 catgcaccta ccagctggcc agaggcatgg agtacttggc ttcccaaaaa tgtattcatc 2521 gaqatttagc agccaqaaat gttttggtaa cagaaaacaa tqtgatgaaa ataqcagact 2581 ttggactcgc caqagatatc aacaatatag actattacaa aaagaccacc aatgggcggc 2641 ttccagtcaa gtggatggct ccagaagccc tgtttgatag agtatacact catcagagtg 2701 atgtctggtc cttcggggtg ttaatgtggg agatcttcac tttagggggc tcgccctacc 2761 cagggattcc ccjtggaggaa cttt taagc tgctgaagga aggacacaga atggataagc 2821 cagccaac g caccaacgaa ctqtacatqa tgatqaggga ctgttggcat gcagtgccct 2881 cccagaqacc aacgttcaag cagtfcggtag aagacttgqa tcgaattctc actctcacaa 2941 ccaatgagga atacttggac ctcagccaac ctctcgaaca gtattcacct agttaccctg 3001 acacaagaag ttcttgttct tcaggagatg attctgtttt ttctccagac cccatgcctt 3061 acgaaccatg ccttcctcacj tatccacaca taaacggcag tgttaaaaca tgaatgactg 3121 gtctgcc g tccccaaaca gqacagcact gggaacctag ctacactqag cagqqagacc 3181 atgcctccca gaqcttgttg tctccacttg tatatatgqa tcagaggagt aaataat gg 3241 aaaagtaatc agcatatgtg taaagattta tacagttgaa aacttgtaat cttccccagg 3301 acjgagaagaa ggtttctgga cjcagtggact gccacaagcc accatgtaac ccctctcacc 3361 tgccgtgcgt actggctgtg gaccagtagg actcaagcjtg gacgtgcgtt ctgccttcct 3421 gttaat t gtaa aattg gaqaagattt atgtcagcac acacttacag agcacaaatg 3481 cagtatatag g tqctggatg tatgtaaata tattcaaatt atg tataaat atatattata 3541 tatttacaag gagttatttt ttgtattgat tttaaatgga tgtcccaatg cacctagaaa 3601 attggtctct ctttttttaa tagctatttg cta.aatgctg ttcttacaca taatttctta 3661 attt caccg agcagagg q gaaaaatact tttgctttca gggaaaatgg tataacgtta 3721 a ttattaat aaattqqtaa tatacaaaac aattaatcat ttatagtttt ttttqtaatt 3781 taagtggcat ttctatgcag gcagcacagc agactagtta atctattgct tqgacttaac 3841 tagttatcag atcctttgaa aagagaatat ttacaatata tgactaattt ggggaaaatg 3901 aagttttgat ttatttgtgt ttaaatgctg ctgtcagacg attgttctta gacctcctaa 3961 atgccccata ttaaaagaac tcattcatag gaaggtgttt cattttggtg tgcaaccctg 4021 cattacgtc aacgcaacgt ctaactggac ttcccaagat aaatggtacc agcgtcctct 4081 taaaagatgc cttaatccat tccttqagga cagaccttag ttgaaatgat aqcagaa gt 4141 gcttctctct ggcagctggc cttctgcttc tgagttgcac attaatcaga ttagcctgta 4201 ttctcttcag tgaattttga taatggcttc cagactcttt ggcgttggag accjcctgtta 4261 ggatc tcaa gtcccatcat agaaaattga aacacagaqt tgttctgctg atagttttgg 4321 ggatacgtcc atctt ttaa ggqattgc t tcatctaatt ctggcaggac ctcaccaaaa 4381 gatccagcct catacctaca tcagacaaaa tatcgccgtt gttccttctg tactaaagta 4441 ttgtgttttg ctttggaaac acccactcac tttgcaatag ccgtgcaaga tgaatgcaga 4501 ttacactgat cttatgtgtt acaaaattcjg agaaagtatt taataaaacc tgttaatttt 4561 tatactgaca ataaaaatgt ttctacagat attaatgtta acaaqacaaa ataaatgtca 4621 cgcaactta ttttfctaata aaaa
100144] The Genbank ID for the TACC2 gene is 10579, Four isoforms are listed for TACC2, e.g., having Genebank Accession Nos. NP_996744 (corresponding nucleotide sequence NM 206862); NP 996743 (corresponding nucleotide sequence NM 206861); NP 996742 (corresponding nucleoiide sequence NM 206860); NP 008928 (corresponding nucleotide sequence NM_ 006997),
[00145] SEQ ID NO: 154 is the TACC2 Amino Acid Sequence for isoform a, having Genebank Accession No. NP_996744 (2948 aa):
1 MGNE 3TSDN QRTLSAQTPR SAQPPGNSQN IKRKQQDTPG SPDHRDASSI GSVGLGGFCT 61 ASESSASLDP CLVSPEVTEP RKDPQGARGP EGSLLPSPPP SQEREKPSSS MPFAECPPEG 121 CLASPAAAPE DGPQTQSPRR EPAPNAPGD1 AAAFPAERDS STPYQETAAV PSAGRERQPK 1 81 E E G Q K S S F S F SSGIDQSPGM SPVPLREPMK APLCGEGDQP GGFESQEKEA AGGFPPAESR 241 QGVASVQVTP EAPAAAQQGT E3SAVLEK3P LKPiiAPIPQD PAPRASDRER GQGEAPPQYL 301 TDDLEFLRAC HLPRS SGAA PEAEV AASQ ESCQQPVGAY LPHAELP GL PSPALVPEAG
361 GSGKEALDTI DVQGHPQTG RGTKPNQWC A G G Q P E G G LPVSPEPSLL T PT E E AH PAS 421 SLASFPAAQ1 PIAVEEPGSS SRESVSKAGM PVSADAAKEV VDAGLVGLER QVSDLGSKGE
481 HPEGDPGEVP APSPQERGEH LNTEQSHEVQ PGVPPPPLPK EQSHEVQPGA PPPPLPKAPS 541 ESARGPPGPT DGAKVHEDST Ξ PAVAKE GSR SPGDSPGGKE EAPEPPDGGD PGNLQGEDSQ 601 AFSSKRDPEV GKDELSKPSS DAESRDHPSS HSAQPPRKGG AGHTDGPHSQ TAEADASGLP 661 KKLGEEDPVL PPVPDGAGEP TVPEGAIWEG SGLQPKCPDT LQSREGLGRM SSFLTLESFK 721 SDFPPTPVAE VAPKAQEGES TLEIRKMGSC DGEGLLTSPD Q P R G P.AC DA S RQEF.HAGVPH 781 PPQGE LAAD LGLTALILDQ DQQGIPSCPG EGWIRGAASE PLLSSEKHL QPSQAQPE S 841 IFDVLKEQAQ PPEKGKETSP SHPGFKDQGA DSSQIHVPVE PQED LPTH GGQEQALGSE 901 LQSQLPKGTL SDTPTSSPTD MVi'JESSLTEE SELSAPTRQK LPALGEKRPE GACGDGQSSR 961 VSPPAADVLK DFSLAGNFSR KETCCTGQGP N K S Q Q AL ADA LEEGSQHEEA CQRHPGASF.A
1021 ADGCSPLWGL S KREMAS G T GEAPPCQPDS VALLDAVPCL PALAPASPGV TPTQDAPETE 1081 ACDETQEGRQ QPVPAPQQKM E C WAT S DAE S PKLLASFPSA G Ξ Q G G Ξ .G AA Ξ G G SAG AG D 1141 PGKQQAPEKP GEATLSCGLL QTEHCLTSGE EASTSALRES CQAEHPMASC QDALLPAREL 1201 GG 1 PRSTMDF S T H Q A P D P K ELLLSGPPEV A AP DT P Y L H DSAAQRGAE D SGVKAVSSAD 12 61 PRAPGESPCP VGEPPLALEN AASLKLFAGS LAPLLQPGAA GGE1PAVQAS SGSPKARTTE 1321 GPVDS PCLD R PLLAKGKQ A G E E K ΑΑΪ P GAG AK S G E GMAGDAAGET EGSMER GEP 1381 SQDPKQGTSG GVDTSSEQIA TLTGFPDFRE HIAKIFEKPV L GAL A P G Ξ K AGAGRSAVGK 1441 DLTRPLGPEK LLDGPPGVDV TLLPAPPARL QVEKKQQLAG EAEISHLALQ DPASDKLLGP 1501 AGLTKER LP GAGVGKSMAG VPPTLREDER PEGPGAAWPG LSGQAYSQLE RSRQSLASGL 1561 PSPAATQELP E R A.AF Q VA P H S H G E E A V A QDRT.PSGKQH Q E S AC D S P H GEDGPGDFAH 1621 TGVPGHVPRS TCAPSPQREV LTVPEANSEP WTLDTLGGER RPGVTAG LE R ALG QST 1681 PAPPTGEVAD T PLE PGKVAG AAGEAEGDIT LSTAETQACA SGDLPEAGTT RTF SWAGDL 1741 VLPGSCQDPA CSDKAPGMEG TAALHGDSPA RPQQAKEQPG PERPIPAGDG KVCVSSPPEP 1801 DBTHDPKLQH LAPEELHTDR ESPRPGPSML PSVPKKDAPR VMDKVTSDST RGAEGTESSP 1861 VADDI IQPAA PADLESPTLA. AS. Ξ Y H G DWG QVSTDLIAQS ISPAAAHAGL PPSAAEHIVS 1921 PSAPAGDRVE ASTPSCPDPA KDLSRSSDSE EAFETPESTT PVKAPPAPPP PPPEVIPEPE 1981 VSTQPPPEEP GCGSETVPVP DGPRSDSVEG SPFRPPSHSF SAVFDEDKPI ASSGTY LDF 2041 DNIELVDTFQ TLEPRASDAK NQEGKVKTRR KSTDSVPISK STLSRSLSLQ ASDFDGASSS 2101 G PSAVALAP DAYSTGSSSA SSTLKRTKKP RPPSLKKKQT TKKPTETPPV KETQQEPDEE 21 61 SLVPSGEKLA SETKTESAKT EGPSPALLEE TPLEPAVGPK AACPLDSESA EGWPPASGG 2221 GRVQKSPPVG RKTLPLTTAP EAGEVTPSDS GGQEDSPAKG LSVRLEFDYS EDKSSWDNQQ 2281 E PPPTKKIG KKPVAKMPLR RPKMKKTPEK LDKTP7 SPPR SPAEPKDIPI AKGTYTFDID 2341 KWDDP F PF SSTSKMQESP KLPQQSYNFD PDTCDESVDP FKTSSKTPSS PSKSPASFEI 2401 PAS AME A G V DGDGLNKPAK KKKTPLKTDT FRVKKSPKRS PLSDPPSQDP TPAA.TPETPP 24 61 VISAWHATD EEKLAVTNQK WTCMTVDLEA DKQDYPQPSD LSTFVKETKF SSPTEELDYR 2521 SYEIEYMEK IGSSLPQDDD APKKQALYLM FDTSQESPVK SSPVEMSESP TPCSGSSFEE 2581 TEALVNTAAK KQHPVPRGLA PNQESHLQVP EKSSQKELEA MGLGTPSEAI E ITAPEGSFA 2641 SADALLSRLA HPVSLCGALD YLEPDLAEK PPLFAQKLQE ELEFAIMRIE ALKLARQIAL 2701 ASRSKQDAKR EAAHPTDVSI SKTALYSRIG TAEVEKPAGL LFQQPDLDSA L Q I A AE I I 2761 KEREVSEBKD K YE E S RRFA/M EMRKIVAEYE KTIAQMIEDE QREKSVSHQT VQQLVLEKEQ 2821 ALADL SVEK SLADLFRRYE KMKEVLEGFR KNEEVLKRCA QEYLSRVKKE EQRYQALKVH 2881 AE E KL DRA A EIAQVRGKAQ QEQAAHQASL RKEQLRVDAL ERTLEQKNKE IEELTKICDE 2941 LIAKMGKS
{00146] SEQ ID NO: 155 is the TACC2 Nucleotide Sequence for isoform a, having Genebank Accession No. NM 206862 (9706 bp)
1 gcctgctcca agggaaggat caggagagaa gaaacgcaaa tcccagaacc gtgccaacat 61 ataaaacccc acattaaggg ttg acagtg cactgggatt tc caagtca cccgcttggt 121 cc bcttccaa gtatacttta c btcctt tca ttcc bctcta aaactt t btt aaaaactttc 181 actcc tgctc taaaagtta t cttggtttct tactctacct tatgcccctt gggcgaattt 241 tttcctctga ggagggaaga atagagttgc tgctgcagac acatcagatt ccctactggt 301 aacagctgga gtgcgtcacc tctgacaaaa ttctggggac gctcjcjgaaca ctgaatcaac 361 atgggcaatg agaacagcac ctcggacaac cagaggactt tatcagetea gactccaagg 421 tccgcgcagc cacccgggaa cag cagaat ataaaaagqa agcagcagga cacgcccgga 481 agccctgacc acagagacgc g tccagcatt ggcaqcqttg gg tggagg cttctgcacc 541 gcttctgaga gttctgccag cctggatcca tgccttgtgt ccccaqaggt gactgagcca 601 aggaaggacc cacagggacjc cagggggeca gaaggttctt tgctgcccag cccaccaccg 661 cccaggagc gagagcaccc ctcgt.ce cc atgccctttg ccgagtgtcc cccgqaaggt 721 tgcttggcaa g tccagcagc ggcacctgaa gatggtcctc agactcagtc tcccaggagg 781 gaacctgccc caaatgcccc agqaqacatc gcggcqqcat ttcccgctga gaggqacagc 841 tctactccat accaagagat tgctgccqtc cccagtgctq gaagaqagag acagecgaaq 901 gaagaaggac acjaagtcctc cttctccttc tccagtggca tcgaccagtc acctggaatg 961 cgccagtac ccc cagaga gecaatgaaq gcaccgctgt gtggagaggg ggaccagcct 1021 ggtggfctttg agtcccaaga gaaagaggct gcaggtggct ttccccctgc aqagtccagg 1081 cagggggtgg cttctqtqca aqtqacccct gaggcccctg ctqcagccca gcagggcaca 1141 gaaagctcag cgqtcttgga qaagtccccc ctaaaaccca tggccccgat cccacaaqat 1201 ccagccccaa gagcctcaga cagacjaaaga ggccaagggg aggcgccgcc tcagtattta 1261 acagatgact tggaattcct caqggcctgc catctcccta ggagcaattc aggggctgee 1321 ccagaagcag aaqtgaatgc egctteccag gagagctgee ageaqecagt qqgagcatat 1381 ctgccgcacg cagagctgcc ctgqqgcttq ccaagtcctg ccctggtgcc agagqctggg 144 ggctctggga aggaggctct cjcjacaccatt gatgttcagg gtcacccaca gacagggatg 1501 cgaggaacca acjcccaatca agttgtctgt gtggcagcag gcggccagcc cgaaggcjcjgt 1561 qcctg ga gccctgaacc ttccctgctc a ccgactg aggaagcaca tccaqcttca 1621 agcctcgctt cattcccagc tgctcagatt ectattgetg tagaagaacc tqgatcatca 1681 tccaqqgaat cagtttccaa ggctqggatq ccagtttctg caqatgcaqc caaagaqgtg 174 gtggatgcag ggttgqtggg actggagagg cagcjtgtcaq atcttggaag caagggagacj 1801 catccagaag gggaccctgg agaggttc t gccc a cac cccaggagag gggagagcac 1861 gaacacgq agcaaagcca tgaggtccaa ccaggagtac caccccctcc tcttcccaag 1921 gagcaaagcc atqaggtcca accagqagca ccaccccetc ctcttcccaa qqcaccaaqt 1981 gaaagtgcca gagggccacc ggggccaacg gatggageca agqtccatqa agattccaca 2041 acjcccagccc; tggctaaaga aggaagcaga tcacctggtg acagccctgg aggaaaggag 2101 gaagccccag agccacctga tggtggagac ccagggaacc tgca ggaga ggactctcag 2161 gctttcagca gcaagcgtga tccagaagta ggcaaagatg aqctttcaaa gecaagcagt 2221 gatgcaqaga gcagagacca tcccaqctca cactcagcac agecacccac! aaaggggqqt 2281 gctgqqcaca cqaacqqqcc ccactctcaq acagcaqagg ctgatgeate tggcctacca 2341 cacaagctcjc; gtgaggagga ccccgtcctg ccccctgtgc cagatggagc tggtgagccc 2401 actg tcccg aaggagccat ctgggagggg eagga qe agcccaaatg tcctgacacc 2461 cttcagagca gggaaqqatt gqqaagaatq gagtctttcc tqactttaqa atcaqagaaa 2521 tcagafctttc caccaactcc tgttgcagag g ttgcaccca aagcccagga aqgtgagagc 2581 acattggaaa taaggaaqat ggqcagctqt gatgqqqagg gettgetqac gtccccagat 2641 caaccccgcc; ggccggcgtg tgatgegteg agacaggaat ttcatgctgg ggtgccacat 2701 cccccccagg gggagaactt ggcagcagac ctggggctca cggcactca cctggaccaa 2761 gatcagcagg gaatcccatc ctqcccagqq gaagqctgga taagaggaqc tgcatccgag 2821 tggcccctac tatcttctga gaagcatctc caqccatccc aggcacaacc agagacatcc 2881 atctttgacq tqctcaaqqa gcaqqcccaq ccacctqaaa atqqgaaaqa gacttctcca 2941 acjccatccac; gttttaagga ccagggacjca gattcttccc aaatccatgt acctgtggaa 3001 cctcaggaag ataacaactt gcccactcat ggaggacagg agcaggcttt gggatcagaa 3061 cfctcaaagfcc agctccccaa aqqcaccctq tctgatactc caacttcatc tcccactgac 3121 atggtfctggg agagttctct gacagaagag tcagaattqt cac!caccaac qagacagaag 3181 ttgcctgcac taggggaqaa gcgqccagaq qgagcatqcg gtqatggtca gtcctcgagg 3241 gtctcgcctc cagcacjcaga tgtcttaaaa gacttttctc ttgcaqcjcjaa cttcagcaqa 3301 aaggaaactt gctgcac gq geaggggeca aacaag ctc aacaggcatt ggctgatgee 3361 tfcggaagaag gcagecagea tqaagaagca tgtcaaaggc atccaggaqc ttctqaagca 3421 gctgatqqtt gttccccact ctggggcttg agtaagaggg agatggcaag tgqaaacaca 3481 ggggaqgccc caccttgtca gcctqactca qtagctctcc tqqatgcaqt tccctqcctg 3541 ccagccc gg cgcccqccag ccccggagtc acacccaccc aqgatgcccc agagacagag 3601 gcatgtgatg aaacccagga aggcaqgeag caac ag qc cggccccgca qcagaaaatg 3661 gagtgctggg ccacttcgga tqeagagtec ccaaaqcttc ttgcaag ttt cccatcagct 3721 gqggagcaag gtgqtgaagc eggggctget gagactggtg geagegctgg tgeaggagae 3781 ccaqqaaagc aqcagqctcc ggaqaaacct qgagaaqcta ctttgagttg tggcctcctt 3841 cagactgagc actqccttac etceggggag gaacjettcta cctctqccct accjtgagtcc 3901 tgccaagctg agcaccccat ggccagctgc caggatgect tgctgccagc cagagagctg 3961 ggtgggattc ccaggagcac catgcjatttt tctacacacc aggctgtccc agacccaaag 4021 gagctcc gc tgtctgggcc accagaagtg gctgctcctg acacccctta ccbgcatgtc 4081 gacagtgctg cccagagagg agcagaagac agtggagtga aagctgtttc ctctgcagac 4141 cccagagckc ctggcgaaag cccctgtcct gtaggggagc ccccacbbcic ctbggaaaab 4201 gctgcctcct tgaagctgtt tgctggctcc ctcgcccccc tgttgcaacc aggagctgca 4261 ggtggcjgaaa tccctgcagt gcaagccagc agtggtacjtc ccaaagccag aaccactcjag 4321 ggaccag gg actccatgcc atgcctggac cggatgccac ttctggccaa gggcaagcag 4381 gcaacagggg aagagaaagc agcaacagct ccaggtgcag gtgccaaggc cagtggggag 4441 ggcatggcag gtgatgcagc aggagagaca gagggcagca tggagaggat gggagagcct 4501 tcccaggacc caaagcaggg cacatcaggt ggtgtggaca caagctctga gcaaatcgcc 4561 accctcactg gcttcccacja cttcagggag cacatcgcca agatcttcga gaagcctcjtg 4621 c cggagccc tggccacacc bggagaaaag gcaggagctg ggaggagtgc agtgggtaaa 4681 gacctcacca ggccattggg cccagagaag cttctagatg ggcctccagg agtggabgtc 4741 acccttctcc ctgcacctcc tgctcgactc caggtggaga agaagcaaca gttggctgga 4801 gaggctgaga tttcccatct ggctctgcaa gatccagctt cagacaagct tctgggtcca 4861 gcaggcjctga cctgggagcg gaacttgcca ggtgccggtg tggggaagga gatggcaggt 4921 g cccaccca cac gaggga agacgagagg ccagaggggc ctggggcagc ctggccaggc 4981 ctggaaggcc aggcttackc acagctggag aggagcaggc aggaattagc ttcaggbctt 5041 ccttcaccag cagctactca ggagctccct gtggagagag ctgctgcctt ccaggtggct 5101 ccccatagcc atggagaaga ggccgtggcc caacjacagaa ttccttctgg aaagcagcac 5161 caggaaacat ctgcctgcga cagtccacat cjcjagaagatg gtcccgggga ctttgctcac 5221 acagggg c caggacatgt gccaaggtcc acgtgtgccc cttctcctca gagggaggtt 5281 ttgacfcgtgc ctgaggccaa cagtgagccc bggacccbtg acacgcttgg gggtgaaagg 5341 agacccggag tcactgctgg catcttggaa atgcgaaatg ccctgggcaa ccagagcacc 5401 cctgcaccac caactggaga acjtggca.cjac actcccctgg agcctggcaa ggtggcaggc 5461 gctgctgggg aagcagaggg tgacatcacc ctgagcacag ctgagacaca ggcatgtgcg 5521 ccggtga c tgcctgaagc aggtac acg aggacattct ccgttgtggc aggtgacttg 5581 gtgctgccag gaagctgtca ggacccagcc bgctctgaca aggctccggg gatggagggt 5641 acagctgccc ttcatgggga cagcccagcc aggccccagc aggctaagga gcagccaggg 5701 cctgagcgcc ccattccagc tggggatggg aagcjtgtgcg tctcctcacc tccagagcct 5761 gacgaaactc acgacccgaa gctgcaacat ttggctccag aagagctcca cactgacaga 5821 gagagcccca ggcctggccc atccatgbta ccttcggttc ctaagaagga tgctccaaga 5881 gtcatggata aagtcactkc agatgagacc agaggtgcgg aaggaacaga aagttcacct 5941 gtggcagatg atatcatcca gcccgctgcc cccgcagacc tggaaagccc aaccttagct 6001 gcctcttcct accaccjgtga tcjttgttcjcjc cagcjtctcta cggatctgat agcccagagc 6061 atctccccag ctgctgccca tgcgggtctt cctccc cgg tgcagaaca catag btcg 6121 ccatctgccc cagcfcggtga cagagtagaa gctbccactc cctcctgccc agatccggcc 6181 aaggacctca gcaggagttc cgatfcctgaa gaggcatbtg agaccccgga gtcaacgacc 6241 cctgtcaaag ctccgccagc tccaccccca ccaccccccg aagtcatccc agaacccgag 6301 g cagcacac agccaccccc cjcjaagaacca ggatgtggtt ctgagacagt ccctgtccct 6361 gatggcccac ggagcgactc ggtggaagga agtcccttcc gtcccccgtc acactccttc 6421 tctgccgtct tcgakgaaga caagccgaba gccagcagtg ggacttacaa ctbggacttb 6481 gacaacattg agcttgtgga tacctttcag accttggagc ctcgtgcctc agacgctaag 6541 aatcaggagg gcaaagtgaa cacacggagg aagtccacgg attccgtccc catctctaag 6601 tctacactcjt cccggtcgct cagcctgcaa gccacjtgact ttgatggtgc ttcttcctca 6661 ggcaa cccg aggccgtggc c t gccc a gatgcatata gcacgggttc cagcagtgct 6721 tctagtaccc ttaagcgaac baaaaaaccg aggccgcctt ccttaaaaaa gaaacagacc 6781 accaagaaac ccacagagac ccccccagtg aaggagacgc aacaggagcc agatgaagag 6841 agccttgtcc ccagtgggga gaatctagca tctgagacga aaacggaatc tgccaagacg 6901 gaaggtccta gcccagcctt attggagcjag acgccccttg agcccgctgt ggggcccaaa 6961 gctgcctgcc ctctggactc agagagtgca gaaggggttg tocccccggc ttctggaggt 7021 ggcagagtgc agaac cacc ccctgtcggg aggaaaacgc bcicctcbbac cacggccccg 7081 gaggcagggg aggtaacccc atcggatagc ggggggcaag aggactctcc agccaaaggg 7141 ctctccgtaa ggctggagtt tgactattct gaggacaaga gtagttggga caaccagcag 7201 gaaaaccccc ctcctaccaa aaagataggc aaaaagccag ttgccaaaat gcccctgagg 7261 aggccaaaga gaaaaagac acccgagaaa cttgacaaca ctcctgcctc acctcccaga 7321 tcccctgcfcg aacccaatga catccccabt gctaaaggta cttacacctt tgatattgac 7381 aagtgggatg accccaattt taaccctttt tcttccacct caaaaatgca ggagtctccc 7441 aaactgcccc aacaabcata caactttgac ccagacacct gtgatgagtc cgttgacccc 7501 tttaagacat cctctaagac ccccagctca ccttctaaat ccccagcctc ctttgagatc 7561 ccagccagtg cbatggaagc caatggagtg gacggggatg ggcbaaacaa gcccgccaag 7621 aagaacjaaga cgcccctaaa gactgacaca tttagggtga aaaacjtcgcc aaaaeggtet 7681 cctctctctg atccaccttc ccaggacccc accccagctg ctacaccaga aacaccaeca 7741 gtga ctctg cggtggtcca cgccacagat gaggaaaagc tggeggtcac caaccagaag 7301 tggacgtgca tgacagtgga cctagaggct gacaaacagg actacccgca gccctcggac 7861 ctgtccacct ttgtaaacga gaccaaattc agttcaccca ctgaggagtt ggattacaga 7921 aactcctatg aaattgaata tatgejagaaa attggctcct ccttacctca ggacgacgat 7981 gccccgaaga ageaggcett gtacct atg tt gacact ctcaggagag ccctgtcaag 8041 teatetcccg tccgcatgtc agagtccccg acgccgtgtt cagggtcaag ttttgaagag 8101 actgaagccc ttgtgaacac tgctgcgaaa aaccagcatc ctgtcccacg aggactggee 8161 cctaaccaag agtcacactt gcaggtgcca gagaaatcct cccagaagga getggaggee 8221 atgggcttgg gcaccccttc agaagegatt cjaaatta.cag ctcccgaggg ctcctttcjcc 8281 etgetgacq ccc cctcag caggctagct caccccgt.c ctctc gtgg tgeacttgae 8341 tatctggagc ccgacttagc agaaaagaac cccccactat tegc cagaa actccaggag 8401 gagttagagt ttgecatcat gcggatagaa gccctgaagc tggecaggea gattgetttg 8461 gcttcccgca gccaccagga tgecaagaga gaggctgetc acccaacaga cgtctccatc 8521 tccaaaacag ccttgtactc ccgcatcggg acegctgagg tggagaaacc tgeaggcett 8581 c gttccagc agcccgacct ggactc gcc ctccagatcg ccagagcaga ga cataacc 8641 aaggagagag aggtctcaga atggaaagat aaatatgaag aaagcaggcg ggaagtgatg 8701 gaaatgagga aaatagtggc cgagtatgag aagaccatcg ctcagatgat agaggacgaa 8761 cagagagacja agtcagtctc ccaccagacg gtgeagcage tggttctgga gaaggagcaa 8821 gccctggccg acctgaactc cgtgcjagaag tctctggccg acctcttcag aagatatgag 8881 aagatgaagg agg cctaga aggcttccgc aagaatgaag aggtg tgaa gagatgtgcg 8941 caggagtacc tgtcccggg t gaagaaggag gagcagaggt accaggccct gaaggtgcac 9001 geggaggaga aactggacag ggecaatget gagattgctc aggttcgagg caaggcccag 9061 caggagcaag ccgcccacca cjgccagcctg eggaaggage agetgegagt ggacgccctg 9121 gaaaggaege ggagcagaa gaa aaagaa atagaagaac tcaccaaga ttgtgacgaa 9181 c gattgeca aaa ggggaa aagctaactc tgaaccgaa gttttggact taactgttgc 9241 gtgcaatatg accgtcggca cactgctgtt cctccagttc catggacagg ttctgtt tc 9301 actttttcgt atgcactact gtatttcctt tctaaataaa attgatttga ttgtatgcag 9361 tactaaggac; actatcagaa tttcttgeta ttggtttgca ttttcctagt ataattcata 9421 gcaagttgac ctcagagttc ctgtatcagg cjagattgtct gattctctaa taaaagacac 9481 a tgetgace ttggccttgc cctttg aca caagttccca gggtgagcag ct ttgga 9541 taatatgaac atgtacagcg tgcataggga c cttgcctt aaggagtgta aacttga ct 9601 geatttgetg atttgttttt aaaaaaacaa gaaatgcatg tttcaaataa aattctctat 9661 tcjtaaataaa attttttctt tggatcttgg caaaaaaaaa aaaaaa
100147] SEQ ID NO: 158 is the amino acid sequence of the FGFR3-TACC3- 1 fusion protein. The bolded text corresponds to the FGF3 protein:
MGAPACALALCVAVAIVAGASSESLGTEQRWG aAEVPGPEPGQQEQLVFGSGDAVSLSCPPPGGGPMGPTVWV DGTGLVPSERrLVGPQRLQVLmSHEDSGAYSCRQRLTQRVLCHFSVRV DAPSSGDDEDGEDEAEDTGVDTGA
PYKTRPERMD KLLAWAAHTTOFRCPAAGNPTPSISWLKHGRSFRGEHRIGGIKLRHQQ SLVMES PSDRGH
YTC EN FGSIRQTYTLDVLERSPHRPILQAGLPANQTAVLGSDWFHC VYSDAQPHXQ L WEWGS VGP
DGTPYV \rLKTAGANTTD ELEVLSLHm^FEDAGEYTCLAGNSIGFSHHSAWLWLPAEEELVEADEAGSVYAG
ILSYGVGFFLFILWAAV LCRLRSPP GLGSPTVH ISRFPL RQVSLESNASMSSiJ PLVRIARLSSGEGPT
LAOTSELELPABP WELSRARLTLGKPLGEGCFGQVVMASAXGIDKDRA& PV VAV MLKDDATD BLSDLVSE
MEM MIGKH HIINLLGACTQGGPLYVLVEYAAKGNLREFLRARRPPGLDYSFDTCKPPEEQLTF DLVSCAYQ
VARGMEYIJ^Q ClHRDLAARNVLWEDNVM IADFGLARDVHNLDYY TT GRLPVKlSmPEA^
DWSFGVLLWEIFTLGGSPYPGIPVEELFKLL EGHRiffi PANCTHDLYMIMRECTHAAPSQRPTFKQLVEDLDR
VLTV STDFKESALRKQSLYLKFDPLLRDSPGRPVPVATETSSMHGA STPSGRPRSAKLVEFDFLGALDIPVPG
PPPGVPAPGGPPLSTGPIVDLLQYSQKDLDAWKATQSENRELRSRCEELHGKNLELGKIMDRFESWYQA EEV
QKQKELSKAEIQKv'LKEKDQLTTDL S EKSFSDLFKRFEKQKEVIEGYRKNEESLKKCVEDYLARITQEGQRYQ
ALKAHAEEKLQLANEE1AQVRSKAQAEALALQASLRKEQ RIQSLEKTVEQKTKENEELTRICDDL1SK EK1
[00148] SEQ ID NO: 159 is the amino acid sequence of the FGFR3 -TACC3 -2 fusion protein. The boided text corresponds to the FGF3 protein: MG&PACAIJ^CVAVAIVAGASSSSLGTEQRWGR&M!VPGPEPGQQEQLVFGSGDAVSLSCPPPGGGPMGPTWV'
KDGTGLVPSERVLVGPQRLQVL ASHEDSGAYSCRQRLTQRVLCHFSVRVTDAPSSGDDEDGEDEAEDTGVDTGA
PYWTRPER J KLLATOAANTVRFRCPAAGNPTPSISWLKNGRSFRGEHRIGGI LRHQQ^SLVMESWPSDRGlSi
YTCWEN FGSIRQTYTLDVLERSPHRPILQAGLPANQTAVLGSDVSFHC WSDAQPHIQWL HV^WGSKVGP
DGTPYV^VL TAGAMTTDKELEVLSLHHV FEDAGEYTCIAGNSIGFSHHSAWLWLPAEEELVEADEAGSVYAG
ILSYGVGFFLFILWAAVTLCRLRSPP GLGSPTVHKXSRFPL RQVSLES ASMSSNTPLVRIARLSSGEGPT
LAWSELELPABPKWELSRARLTLGKPLGEGCFGQVV^^
MSMdMIGKHKiillNLLGACTQG^
VARGMEY]^QKCIHRDLAARf¾WEDfVM I
D SFGVLLWEIFTLGGSPYPGIPVEELFKLLKEGHR KPAHCTKDLYMIMRECWHAAPSQRPTF QLVEDLDR
VLTVTSTD\/SAGSGIA/PPAYAPPPAVPGHPSGRPREAKLVEFDFLGALDIpypGPPPGVPAPGGPPLSTGPIVDL LQYSQKDLDAV\/KATQEE RELRSRCEELHGK LELGKIMDRFEEV\/YQA EE\/QKQKELSKAEIQK\/LKEKDQL TTDLNS EKSFSDLFKRFEKQKEVIEGYRK EESLKKCVEDYIAR TQEGQRYQALKAHAEEKLQLANEEIAQVR SKAQAEALALQASLRKSQMRIQSLSKTVEQKTKE EELTRICDDLISKMEKI
{0014.9] SEQ ID NO: 160 is the amino acid sequence of the FGFR3-TACC3-3 fusion protein. The bokled text corresponds to the FGF3 protein:
MGAPACALALCVAVAIVAGASSESLGTEQRWGRAAEVPGPEPGQQEQLVFGSGDAVELSCPPPGGGPMGPT V DGTGLVPSERVLVGPQRLQVLNASHEDSGAYSCRQRLTQRVLCHFS¥RV DAPSSGDDEDGEDEAEDTGVDTGA PYWTRPER 3 LLAWAANTVRFRCPAAGNPTPSISWL NGREFRGEHRIGGIKLRHQQ SL\¾ESVVPSDRGN YTC\^N FGSIRQTYTLDVLERSPHRPILQAGLPANQTAVLGSD\rEFHC VYSDAQPHIQ L HVEVNGS VGP DGTPYWVL TAGANTTDKELEVLSLH V FEDAGEYTCIAGNSIGFSHHSAWLWLPAEEELVEADEAGSVYAG ILSYGVGFFLFILWAAVTLCRLRSPPKKGLGSPTVH ISRFPLKRQVSLESNASMSSOTPLVRIARLSSGEGPT IJWrSELELPADPKWELSRARLTLGKPLGEGCFGQVV A^
MEiiMKMIGKH NIINLLGACTQGGPLYVLVEYAA Gl&REFLRARRPPGLDYSFDTCKPPEEQLTF DLVSCAYQ VARGMEYLASQKCIHRDL A mrLV EDm^^
DVWSFG\¾LWEIFTLGGSPYPGIPVEELFKLLKEGHRMDKPANCTHDLYMIMRECWKAAPSQRPTFKQLVEDLDR
VLTVTSTDVPGPPPGVPAPGGPPLSTGPIVDLLQYSQKDLDAWRATQSENRELRSRCEELHGKNLELGKIMDRF EEvry"YQAMESVQKQKELSKAEIQKVLKEKDQirTDL SMEKSFSDLFKRFEKQKEVIEGYRKNEESLKKCVEDYL ARITQEGQRYQALKAHAEEKLQLANEEIAQVRSKAQAEALALQASLRKEQ RIQSLEKTVEQKTKENEELTRICD DLISK EKI
[00150] SEQ ID NO: 161 is the amino acid sequence of the FGFR3-TACC3-4 fusion protein. The boided text corresponds to the FGF3 protein:
MGAPACJkLALCVAVAIVAGASSESLGTEQRWGRAAEVPGPEPGQQEQLVFGSGDAVELSCPPPGGGPMGPTVW KDGTGLVPSERVLVGPQRLQVL ASHEDSGAYSCRQRLTQRVLCHFSVRV DAPSSGDDEDGEDEAEDTGVDTGA PYWTRPERMDK LLAVPAANTVRFRCPAAGNPTPSlSWL NGREFRGEHRlGGIKLReQQ SLVMESWPSDRGlSf YTCVVEN FGSIRQTYTLDVLERSPHRPILQAGLPMQTAVLGSDVEFHC V SDAQPHIQWL HV^TOGS VGP DGTPYV VL TAGANTTD ELE LSLHNWFEDAGEYTCIAGNSIGFSHHSAWLWLPAEEELVEADEAGSVYAG ILSYGVGFFLFILWAAV LCRLRSPP GLGSPTVHKISRFPLKRQVSLESNASMSSH PLVRIARLSSGEGPT LANVSELELPADPKWELSRARLTLGKPLGEGCFGQVVMAEAIGID DRAAKPVTVAVKMLKDDATD DLSDLVSE MEiMMlG HK IlNLLGACTQGGPLYVLVEYAAKGNLREFLRARRPPGLDYSFDTC PPEEQLTFKDLVSCAYQ VARGMEYLASQKCIHRDLAAROTLWEDiW^
DWSFGVLLWEIFTLGGSPYPGIPVEELFKLL EGHRMD PANCTHDLYMIMRECWHAAPSQRPTF QLVEDLDR
VLTV STDVKATQEE RELRSRCEELHGK LELGKIMDRFEEVVYQAMEEVQKQKELSKAEIQKVLKEKDQLTTD LNSMEKSFSDLFKRFEKQKEv'IEGYRKNEESLKKCVEDYLARI QEGQRYQALKA.HAEEKLQLANEEIA.QVRSKA QASALALQASLRKEQMRIQSLEKTVEQKTKEKEELTRICDDLISKMEKI
[OOISI] The Genbank ID for the FGF 4 gene is 2.264. Three isoforms are listed for FGF 4, e.g., having Genebank Accession Nos. NP_002002 (corresponding nucleotide sequence NM_002011); NP_075252 (corresponding nucleotide sequence NM_022963); NP 998812 (corresponding nucleotide sequence NM 213647).
{00152] As used herein, a "FGFR fusion molecule" can be a nucleic acid (e.g., synthetic, purified, and/or recombinant) which encodes a polypeptide corresponding to a fusion protein comprising a tyrosine kinase domain of an FGFR protein fused to a polypeptide that constitutively activates the tyrosine kinase domain of the FGFR protein, or a nucleic acid encoding a fusion protein comprising a transforming acidic coiled-coil (TACC) domain fused to a polypeptide with a tyrosine kinase domain, wherein the TACC domain constitutively activates the tyrosine kinase domain. It can also be a fission protein comprising a tyrosine kinase domain of an FGFR protein fused to a polypeptide that constitutively activates the tyrosine kinase domain of the FGFR protein, or a fusion protein comprising a transforming acidic coiled-coil (TACC) domain fused to a polypeptide with a tyrosine kinase domain, wherein the TACC domain constitutively activates the tyrosine kinase domain. For example, a FGFR fusion molecule can include a FGFR 1 -TACC 1 (e.g., comprising the amino acid sequence shown in SEQ ID NO: 150, or comprising the nucleic acid sequence shown in SEQ ID NO: 88), FGFR2-TACC2, FGFR3-TACC3 (e.g., comprising the amino acid sequence shown in SEQ ID NOS: 79, 158-160, or 161, or comprising the nucleic acid sequence shown in SEQ ID NOS: 80-82, 84, 94- 144, or 145), or other FGFR-TACC fusion proteins (e.g., an N-terminai fragment of FGFR4 containing its tyrosine kinase domain fused to a fragment of TACC1, TACC2, or TACC 3). For example, a FGFR fusion molecule can include a FGFR1- containing fusion comprising the amino acid sequence corresponding to Genebank Accession no. NP 001167534, NP 001 167535, NP 001 167536, NP 001 167537, NP 001 167538, NP_056934, NPJ375593, NP_075594, or NPJ375598; or a FGFR 1 -containing fusion comprising the nucleotide sequence corresponding to Genebank Accession no.
NM_001 174063, NM_001 174064, NM 001 174065, NM_001 174066, W) 00 ) 174067, NM__ 015850, NM__ 023105, NM___023106, or NM__0231 10. For example, a FGFR fusion molecule can include a FGFR2-containing fusion comprising the amino acid sequence corresponding to Genebank Accession no. NP_ 000132, NP_001 138385, NP_ 001 138386, NP 001 138387, NP 001 138388, NP 001 138389, NP 001 138390, NP 001 138391, or NP 075259; or a FGFR2 -containing fusion comprising the nucleotide sequence
corresponding to Genebank Accession no. NM 000141 , NM_001 144913, NM 00 i 144914, NM_001 144915, NM 00 i 144916, \ \ 001 14491 7. NM_001 144918, NM OOl 144919, or NM 022970. For example, a FGFR fusion molecule can include a FGFR3 -containing fusion comprising the amino acid sequence corresponding to Genebank Accession no. NP_000133, P 001 156685. or NP 075254; or a FGFR3-containing fusion comprising the nucleotide sequence corresponding to Gene-bank Accession no, NM 000142, NM 001 163213, or NM_022965. For example, a FGFR fusion molecule can include a FGFR4-containing fusion comprising the amino acid sequence corresponding to Genebank Accession no. NP_002002, NP 075252, or NP 998812; or a FGFR4-containing fusion comprising the nucleotide sequence corresponding to Genebanli Accession no. NM 00201 1, NM 022963, or
NM_213647. A FGFR fusion molecule can also include a tyrosine kinase domain of an FGFR protein fused to a protein encoded by any one of the genes listed in Table 7. A FGFR fusion moiecule can include a variant of the above described examples, such as a fragment thereof.
[Θ0153] Table ?. Fusion Partners
[©0154] The nucleic acid cars he any type of nucleic acid, including genomic DNA, complementary DMA (cDNA), recombinant DNA, synthetic or semi-synthetic DNA, as well as any form of corresponding RNA. A cDNA is a form of DNA artificially synthesized from a messenger RNA template and is used to produce gene clones. A synthetic DNA is free of modifications that can be found in cellular nucleic acids, including, but not limited to, histones and methylation. For example, a nucleic acid encoding a FGFR fusion molecule can comprise a recombinant nucleic acid encoding such a protein. The nucleic acid can be a non- naturally occurring nucleic acid created artificially (such as by assembling, cutting, ligating or amplifying sequences). It can be double- stranded or single-stranded.
[0(5155] The invention further provides for nucleic acids that are complementary to a FGFR fusion molecule. Complementary nucleic acids can hybridize to the nucleic acid sequence described above under stringent hybridization conditions. Non-limiting examples of stringent hybridization conditions include temperatures above 30°C, above 35°C, in excess of 42°C, and/or salinity of less than about 500 mM, or less than 200 mM. Hybridization conditions can be adjusted by the skilled artisan via modifying the temperature, salinity and/or the concentration of other reagents such as SDS or SSC. [0(5156] According to the invention, protein variants can include amino acid sequence modifications. For example, amino acid sequence modifications fall into one or more of three classes: substitutional, insertional or deietional variants. Insertions can include amino and/or carboxyl terminal fusions as well as intrasequence insertions of single or multiple amino acid residues. Insertions ordinarily will be smaller insertions than those of amino or carboxyl terminal fusions, for example, on the order of one to four residues. Deletions are characterized by the removal of one or more amino acid residues from the protein sequence. These variants ordinarily are prepared by site-speci fic mutagenesis of nucleotides in the DNA encoding the protein, thereby producing DNA encoding the variant, and thereafter expressing the DNA in recombinant cell culture.
[!>Θ157] In one embodiment, a FGFR fusion molecule comprises a protein or polypeptide encoded by a nucleic acid sequence encoding a FGFR fusion molecule, such as the sequences shown in SEQ ID NOS: 80-82, 84, 94-144, or 145. In some embodiments, the nucleic acid sequence encoding a FGFR fusion molecule is about 70%, about 75%, about 80%, about 85%, about 90%, about 93%, about 95%, about 97%, about 98%, or about 99% identical to SEQ ID NOS: 80-82, 84, 94- 144, or 145. Tn another embodiment, the polypeptide can be modified, such as by glycosylations and/or acetylations and/or chemical reaction or coupling, and can contain one or several non-natural or synthetic amino acids. An example of a FGFR fusion molecule is the polypeptide having the amino acid sequence shown in SEQ ID NOS: 79, 88, 150, 158-160, or 161. In some embodiments, the FGFR fusion molecule that is a polypeptide is about 70%, about 75%, about 80%, about 85%, about 90%, about 93%, about 95%, about 97%, about 98%, or about 99% identical to SEQ ID NOS: 79, 88, 150, 158-160, or 161. In another embodiment, a FGFR fusion molecule can be a fragment of a FGFR fusion protein. For example, the FGFR fusion molecule can encompass any portion of at least about 8 consecutive amino acids of SEQ ID NOS: 79, 88, 150, 158-160, or 161. The fragment can comprise at least about 10 amino acids, a least about 20 amino acids, at least about 30 amino acids, at least about 40 amino acids, a least about 50 amino acids, at least about 60 amino acids, or at least about 75 amino acids of SEQ ID NOS: 79, 88, 150, 158-160, or 161 , Fragments include ail possible amino acid lengths between about 8 and 100 about amino acids, for example, lengths between about 10 and 100 amino acids, between about 15 and 100 amino acids, between about 20 and 100 amino acids, between about 35 and 1 0 amino acids, between about 40 and 100 amino acids, between about 50 and 100 amino acids, between about 70 and 100 amino acids, between about 75 and 100 amino acids, or between about 80 and 100 amino acids. Fragments include all possible amino acid lengths between about 100 and 800 amino acids, for example, lengths between about 125 and 800 amino acids, between about 150 and 800 amino acids, between about 175 and 800 amino acids, between about 200 and 800 amino acids, between about 225 and 800 amino acids, between about 250 and 800 amino acids, between about 275 and 800 amino acids, between about 300 and 800 amino acids, between about 325 and 800 amino acids, between about 350 and 800 amino acids, between about 375 and 800 amino acids, between about 400 and 800 amino acids, between about 425 and 800 amino acids, between about 450 and 800 amino acids, between about 475 and 800 amino acids, between about 500 and 800 amino acids, between about 525 and 800 amino acids, between about 550 and 800 amino acids, between about 575 and 800 amino acids, between about 600 and 800 amino acids, between about 625 and 800 amino acids, between about 650 and 800 amino acids, between about 675 and 800 amino acids, between about 700 and 800 amino acids, between about 725 and 800 amino acids, between about 750 and 800 amino acids, or between about 775 and 800 amino acids.
[00158] Chemical Synthesis. Nucleic acid sequences encoding a FGFR fusion molecule can be synthesized, in whole or in part, using chemical methods known in the art.
Alternatively, a polypeptide can be produced using chemical methods to synthesize its amino acid sequence, such as by direct peptide synthesis using solid-phase techniques. Protein synthesis can either be performed using manual techniques or by automation. Automated synthesis can be achieved, for example, using Applied Biosystems 431 A Peptide Synthesizer (Perkin Elmer).
[00159] Optionally, polypeptides fragments can be separately synthesized and combined using chemical methods to produce a full-length molecule. For example, these methods can be util ized to synthesize a fusion protein of the invention, in one embodiment, the fusion protein comprises a tyrosine kinase domain of an FGFR protein fused to a polypeptide that constitutive ly activates the tyrosine kinase domain of the FGFR protein. In another embodiment, a fusion protein comprises a transforming acidic coiled-coil (TACC) domain fused to a polypeptide with a tyrosine kinase domain, wherein the TACC domain constitutively activates the tyrosine kinase domain. An example of a fusion protein is the FGFR1-TACC1 polypeptide, which comprises the amino acid sequence shown in SEQ ID NO: 150. An example of a fusion protein is the FGFR3-TACC3 polypeptide, which has the amino acid sequence comprising SEQ TD NO: 79, 158, 159, 160, or 161 . [0(5160] Obtaining, Purifying and Detecting FGFR fusion molecules. A polypeptide encoded by a nucleic acid, such as a nucleic acid encoding a FGFR fusion molecule, or a variant thereof, can be obtained by purification from human ceils expressing a protein or polypeptide encoded by such a nucleic acid. Non-limiting purification methods include size exclusion chromatography, ammonium sulfate fractionation, ion exchange chromatography, affinity chromatography, and preparative gel electrophoresis.
[0(5161] A synthetic polypeptide can be substantially purified via high performance liquid chromatography (HPLC), such as ion exchange chromatography (IEX-HPLC). The composition of a synthetic polypeptide, such as a FGFR fusion molecule, can be confirmed by amino acid analysis or sequencing.
[0(5162] Other constructions can also be used to join a nucleic acid sequence encoding a polypeptide/prolein of the claimed invention to nucleotide sequence encoding a polypeptide domain which will facilitate purification of soluble proteins. Such purification facilitating domains include, but are not limited to, metal chelating peptides such as histidine-tryptophan modules that allow purification on immobilized metals, protein A domains that allow purification on immobilized immunoglobulin, and the domain utilized in the FLAGS extension/aftlnity purification system (immunex Corp., Seattle, Wash.), including cleavable linker sequences (i.e., those specific for Factor Xa or enterokinase (Tnvitrogen, San Diego, Calif.)) between the purification domain and a polypeptide encoded by a nucleic acid of the invention also can be used to facilitate purification. For example, the skilled artisan can use an expression vector encoding 6 histidine residues that precede a thioredoxin or an enterokinase cleavage site in conjunction with a nucleic acid of interest. The histidine residues facilitate purification by immobilized metal ion affinity chromatography, while the enterokinase cleavage site provides a means for purifying the polypeptide encoded by, for example, an FGFRl-TACCi, FGFR2-TACC2, FGFR3-TACC3, other FGFR-TACC, FGFR- containing, or TACC containing nucleic acid.
[00163] Host cells which contain a nucleic acid encoding a FGFR fusion molecule, and which subsequently express the same, can be identified by various procedures laiown to those of skill in the art. These procedures include, but are not limited to, DNA-D or DNA-R A hybridizations and protein bioassay or immunoassay techniques which include membrane, solution, or chip-based technologies for the detection and/or quantification of nucleic acid or protein. For example, the presence of a nucleic acid encoding a FGFR fusion molecule can be detected by D A-DNA or DNA-RNA hybridization or amplification using probes or fragments of nucleic acids encoding (he same. In one embodiment, a nucleic acid fragment of a FGFR fusion molecule can encompass any portion of at least about 8 consecutive nucleotides of SEQ TD NOS: 80-82, 84, 94- 144, or 145. In another embodiment, the fragment can comprise at least about 10 consecutive nucleotides, at least about 15 consecutive nucleotides, at least about 20 conseutive nucleotides, or at least about 30 consecutive nucleotides of SEQ ID NOS: 80-82, 84, 94- 144, or 145. Fragments can include all possible nucleotide lengths between about 8 and about 100 nucleotides, for example, lengths between about 1 and about 100 nucleotides, or between about 2.0 and about 100 nucleotides. Nucleic acid amplification-based assays involve the use of oligonucleotides selected from sequences encoding a FGFR fusion molecule nucleic acid, or FGFR fusion molecule nucleic acid to detect transformants which contain a nucleic acid encoding a protein or polypeptide of the same.
[0(5164] Protocols are known in the art for detecting and measuring the expression of a polypeptide encoded by a nucleic acid, such as a nucleic acid encoding a FGFR fusion molecule, using either polyclonal or monoclonal antibodies specific for the polypeptide. Non-limiting examples include enzyme-linked immunosorbent assay (ELISA),
radioimmunoassa (RIA), and fluorescence activated cell sorting (FACS), A two-site, monoclonal-based immunoassay using monoclonal antibodies reactive to two no n- interfering epitopes on a polypeptide encoded by a nucleic acid, such as a nucleic acid encoding a FGFR fusion molecule, can be used, or a competitive binding assay can be employed.
[00165] Labeling and conjugation techniques are known by those skilled in the art and can be used in various nucleic acid and amino acid assays. Methods for producing labeled hybridization or PGR probes for detecting sequences related to nucleic acid sequences encoding a protein, such as FGFR fusion molecule, include, but are not limited to, oligolabeling, nick translation, end-labeling, or PGR amplification using a labeled nucleotide. Alternatively, nucleic acid sequences, such as nucleic acids encoding a FGFR fusion molecule, can be cloned into a vector for the production of an mRNA probe. Such vectors are known in the art, are commercially available, and can be used to synthesize RNA probes in vitro by addition of labeled nucleotides and an appropriate RNA polymerase such as T7, T3, or SP6. These procedures can be conducted using a variety of commercially available kits (Amersham Pharmacia Biotech, Promega, and US Biochemical). Suitable reporter molecules or labels which can be used for ease of detection include radionuclides, enzymes, and fluorescent, ehemilumineseent, or chromogenic agents, as well as substrates, eofactors, inhibitors, and/or magnetic particles.
[00166] A fragment can be a fragment of a protein, such as a FGFR fusion protein. For example, a fragment of a FGFR fusion molecule can encompass any portion of at least about 8 consecutive amino acids of SEQ ID NOS: 79, 88, 150, 158-160, or 161. The fragment can comprise at least about 10 consecutive amino acids, at least about 20 consecutive amino acids, at least about 30 consecutive amino acids, at least about 40 consecutive amino acids, a least about 50 consecutive amino acids, at least about 60 consecutive amino acids, at least about 70 consecutive amino acids, at least about 75 consecutive amino acids, at least about 80 consecutive amino acids, at least about 85 consecutive amino acids, at least about 90 consecutive amino acids, at least about 95 consecutive amino acids, at least about 100 consecutive amino acids, at least about 200 consecutive amino acids, at least about 300 consecutive amino acids, at least about 400 consecutive amino acids, at least about 500 consecutive amino acids, at least about 600 consecutive amino acids, at least about 700 consecutive amino acids, or at least about 800 consecutive amino acids of SEQ ID NOS: 79, 88, 150, 158- 160, or 161. Fragments include all possible amino acid lengths between about 8 and 100 about amino acids, for example, lengths between about 10 and about 100 amino acids, between about 15 and about 100 amino acids, between about 20 and about 100 amino acids, between about 35 and about 100 amino acids, between about 40 and about 100 amino acids, between about 50 arid about 100 amino acids, between about 70 arid about 100 amino acids, between about 75 and about 100 amino acids, or between about 80 and about 100 amino acids.
Ceil Tra fection
[0(5167] Host cells transformed with a nucleic acid sequence of interest can be cultured under conditions suitable for the expression and recovery of the protein from cell culture. The polypeptide produced by a transformed cell can be secreted or contained intracellularly depending on the sequence and/or the vector used. Expression vectors containing a nucleic acid sequence, such as a nucleic acid encoding a FGFR fusion molecule, can be designed to contain signal sequences which direct secretion of soluble polypeptide molecules encoded by the nucleic acid. Cell transfection and culturing methods are described in more detail below. [0(51 8] A eukaryotic expression vector can be used to transfect cells in order to produce proteins encoded by nucleotide sequences of the v ector, e.g. those encoding a FGFR fusion molecule. Mammal ian cells can contain an expression vector (for example, one that contains a nucleic acid encoding a fusion protein comprising a tyrosine kinase domain of an FGFR protein fused to a polypeptide that constitutively activates the tyrosine kinase domain of the FGFR protein, or a nucleic acid encoding a fusion proiein comprises a transforming acidic coiled-coil (TACC) domain fused to a polypeptide with a tyrosine kinase domain, wherein the TACC domain constitutively activates the tyrosine kinase domain) via introducing the expression vector into an appropriate host cell via methods known in the art.
[00169] A host cell strain can be chosen for its ability to modulate the expression of the inserted sequences or to process the expressed polypeptide encoded by a nucleic acid, in the desired fashion. Such modifications of the polypeptide include, but are not limited to, acetylation, carboxyiation, glycosylation, phosphorylation, lipidation, and acyiation. Post- translational processing which cleaves a "prepro" form of the polypeptide also can be used to facilitate correct insertion, folding and/or function. Different host cells which have specific cellular machinery and characteristic mechanisms for post-translational activities (e.g., CHO, HeLa, MDC , HEK293, and WI38), are available from the American Type Culture Collection (ATCC; 10801 University Boulevard, Manassas, Va. 20110-2209) and can be chosen to ensure the correct modification and processing of the foreign protein.
[00170] An exogenous nucleic acid can be introduced into a cell via a variety of techniques known in the art, such as Hpofection, microinjection, calcium phosphate or calcium chloride precipitation, DEAE-dextran-mediated transfection, or electroporation. Electroporation is carried out at approximate voltage and capacitance to result in entry of the DNA constructs) into cells of interest (such as glioma cells (ceil line SF 188), neuroblastoma cells (ceil lines lMR-32, SK-N-SH, SH-F and SFl-N), astrocytes and the like). Other transfection methods also include modified calcium phosphate precipitation, polybrene precipitation, liposome fusion, and receptor -mediated gene delivery.
[00171] Cells that will be genetically engineered can be primary and secondary cells obtained from various tissues, and include ceil types which can be maintained and propagated in culture. Non-limiting examples of primary and secondar '- ceils include epithelial cells, neural cells, endothelial cells, glial cells, fibroblasts, muscle cells (such as myoblasts) keratinocytes, formed elements of the blood (e.g., lymphocytes, bone marrow cells), and precursors of these somatic ceil types.
[!>Θ172] Vertebrate tissue can be obtained by methods known to one skilled in the art, such a punch biopsy or other surgical methods of obtaining a tissue source of the primary cell type of interest. In one embodiment, a punch biopsy or removal (e.g., by aspiration) can be used to obtain a source of cancer cells (for example, glioma cells, neuroblastoma cells, and the like ). A mixture of primary cells can be obtained from the tissue, using methods readily practiced in the art, such as explanting or enzymatic digestion (for examples using enzymes such as pronase, trypsin, collagenase, elastase dispase, and chymotrypsin). Biopsy methods have also been described in United States Patent No. 7,419,661 and PCT application publication WO 2001/32840, and each are hereby incorporated by reference.
[0(5173] Primary cells can be acquired from the individual to whom the genetically engineered primary or secondary cells are administered. However, primary cells can also be obtained from a donor, other than the recipient, of the same species. 'T'he cells can also be obtained from another species (for example, rabbit, cat, mouse, rat, sheep, goat, dog, horse, cow, bird, or pig). Primary cells can also include cells from a purified vertebrate tissue source grown attached to a tissue culture substrate (for example, flask or dish) or grown in a suspension; cells present in an explant derived from tissue; both of the aforementioned cell types plated for the first rime; and cell culture suspensions derived from these plated ceils. Secondary cells can be plated primary cells that are removed from the culture substrate and replated, or passaged, in addition to cells from the subsequent passages. Secondary cells can be passaged one or more times. These primary or secondary cells can contain expression vectors having a gene that encodes a FGFR fusion molecule.
Cell Culturing
[$$174] Various culturing parameters can be used with respect to the host cell being cultured. Appropriate culture conditions for mammalian cells are well known in the art (Cleveland WL, et al. , J Immunol Methods, 1983, 56(2): 223 -234) or can be determined by the skilled artisan (see, for example, Animal Cell Culture: A Practical Approach 2nd Ed., RickwoocL D. and Hames, B. D., eds. (Oxford University Press: New York, 1992)). Cell culturing conditions can vary according to the ty pe of host cell selected. Commercially available medium can be utilized. Non-limiting examples of medium include, for example, Minimal Essential Medium (MEM, Sigma. St. Louis, Mo.); Dulbecco's Modified Eagles Medium (DMEM, Sigma); Ham's F10 Medium (Sigma); HyClone ceil culture medium (ByCione, Logan, Utah); RPMT-1640 Medium (Sigma); and chemically-defined (CD) media, which are formulated for various cell types, e.g., CD-CHO Medium (Invitrogen, Carlsbad, Calif.).
[0(5175] The cell culture media can be supplemented as necessary with supplementary components or ingredients, including optional components, in appropriate concentrations or amounts, as necessary or desired. Cell culture medium solutions provide at least one component from one or more of the following categories: (I) an energy source, usually in the form of a carbohydrate such as glucose; (2) all essential amino acids, and usually the basic set of twenty amino acids plus cysteine; (3) vitamins and/or other organic compounds required at low concentrations; (4) free fatty acids or lipids, for example linoleic acid; and (5) trace elements, where trace elements are defined as inorganic compounds or naturally occurring elements that can be required at very low concentrations, usually in the micromolar range.
[Θ0176] The medium also can be supplemented electively with one or more components from any of the following categories: (1) salts, for example, magnesium, calcium, and phosphate; (2) hormones and other growth factors such as, serum, insulin, transferrin, and epidermal growth factor; (3) protein and tissue hydrolysates, for example peptone or peptone mixtures which can be obtained from purified gelatin, plant material, or animal byproducts; (4) nucleosides and bases such as, adenosine, thymidine, and hypoxanthine; (5) buffers, such as HEPES; (6) antibiotics, such as gentamycin or ampicillin; (7) cell protective agents, for example pluronic polyoi; and (8) galactose. In one embodiment, soluble factors can be added to the culturing medium.
[0(5177] The mammalian cell culture that can be used with the present invention is prepared in a medium suitable for the type of cell being cultured. In one embodiment, the cell culture medium can be any one of those previously discussed (for example, MEM) that is supplemented with serum from a mammalian source (for example, fetal bovine serum (FBS)). In another embodiment, the medium can be a conditioned medium to sustain the growth of host ceils.
[0(5178] Three-dimensional cultures can be formed from agar (such as Gey's Agar), hydrogels (such as matrigei, agarose, and the like; Lee et a!., (2004) Biomalerials 25: 2461- 2466) or polymers that are cross-linked. These polymers cars comprise natural polymers and their derivatives, synthetic polymers and their derivatives, or a combination thereof. Natural polymers can be anionic polymers, cationic polymers, amphipathic polymers, or neutral polymers. Non-limiting examples of anionic polymers can include hyaluronic acid, alginic acid (alginate), carageenan, chondroi tin sulfate, dextran sulfate, and pectin. Some examples of cationic polymers, include but are not limited to, chitosan or poly lysine. (Peppas et al., (2006) Adv Mater. 18: 1345-60; Hoffman, A. S„ (2002) Adv Drug Deliv Rev, 43: 3- 12; Hoffman, A. S., (2001) Ann NY Acad Sci 944: 62-73). Examples of amphipathic polymers can include, but are not limited to collagen, gelatin, fibrin, and earboxymethyl chitin. Non- limiting examples of neutral polymers can include dextran, agarose, or puiluian. (Peppas et al, (2006) Adv Mater. 18: 1345-60; Hoffman, A. S„ (2002) Adv Drug Deliv Rev, 43: 3- 12; Hoffman, A. S., (2001 ) Ann NY Acad Sci 944: 62-73).
[00179] Cells to be cultured can harbor introduced expression vectors, such as plasmids. The expression vector constructs can be introduced via transformation, microinjection, transfection, lipofection, electroporation, or infection. The expression vectors can contain coding sequences, or portions thereof, encoding the proteins for expression and production. Expression vectors containing sequences encoding the produced proteins and polypeptides, as well as the appropriate transcriptional and translalional control elements, can be generated using methods well known to and practiced by those skilled in the art. These methods include synthetic techniques, in vitro recombinant DNA techniques, and in vivo genetic recombination which are described in J. Sambrook et al., 2001, Molecular Cloning, A
Laboratory Manual. Cold Spring Harbor Press, Plainview, N.Y. and in F. M. Ausubel et al., 1989, Current Protocols in Molecular Biology, John Wiley & Sons, New York, N.Y. ft i ' R ft ''usion Molecule inhibitors
[Θ018Θ] The invention provides methods for use of compounds that decrease the expression level or activity of a FGFR fusion molecule in a subject, in addition, the invention provides methods for using compounds for the treatment of a gene-fusion associated cancer. In one embodiment, the gene-fusion associated cancer is an epithelial cancer. In one embodiment, the gene-fusion associated cancer comprises glioblastoma multiforme, breast cancer, lung cancer, prostate cancer, or colorectal carcinoma. [0(5181] As used herein, a "FGFR fusion molecule inhibitor" refers to a compound that interacts with a FGFR fusion molecule of the invention and modulates lis activity and/or its expression. For example, the compound can decrease the activity or expression of a FGFR fusion molecule. The compound can be an antagonist of a FGFR fusion molecule (e.g., a FGFR fusion molecule inhibitor). Some non- limiting examples of FGFR fusion molecule inhibitors include peptides (such as peptide fragments comprising a FGFR fusion molecule, or antibodies or fragments thereof), small molecules, and nucleic acids (such as siRNA or antisense RNA specific for a nucleic acid comprising a FGFR fusion molecule). Antagonists of a FGFR fusion molecule decrease the amount or the duration of the activity of an FGFR fusion protein. In one embodiment, the fission protein comprises a tyrosine kinase domain of an FGFR protein fused to a polypeptide that constitutively activates the tyrosine kinase domain of the FGFR protein (e.g., FGFR 1 -TACC 1, FGFR2-TACC2, FGFR3-TACC3 or other FGFR-TACC), or a fusion protein comprises a transforming acidic coiled-coii (TACC) domain fused to a polypeptide with a tyrosine kinase domain, wherein the TACC domain constitutively activates the tyrosine kinase domain. Antagonists include proteins, nucleic acids, antibodies, small molecules, or any other molecule which decrease the activity of a FGFR fusion molecule.
[00182J The term "modulate," as it appears herein, refers to a change in the activity or expression of a FGFR. fusion molecule. For example, modulation can cause a decrease in protein activity, binding characteristics, or any other biological, functional, or immunological properties of a FGFR fusion molecule, such as an FGFR fusion protein.
[Θ0183] In one embodiment, a FGFR fusion molecule inhibitor can be a peptide fragment of a FGFR fusion protein that binds to the protein itself.
[00184] For example, the FGFR fusion polypeptide can encompass any portion of at least about 8 consecutive amino acids of SEQ ID NOS: 79, 88, 150, 158- 160, or 161. The fragment can comprise at least about 10 consecutive amino acids, at least about 20 consecutive amino acids, at least about 30 consecutive amino acids, at least about 40 consecutive amino acids, a least about 50 consecutive amino acids, at least about 60 consecutive amino acids, at least about 70 consecutive amino acids, at least about 75 consecutive amino acids, at least about 80 consecutive amino acids, at least about 85 consecutive amino acids, at least about 90 consecutive amino acids, at least about 95 consecutive amino acids, at least about 100 consecutive amino acids, at least about 200 consecutive amino acids, at least about 300 consecutive amino acids, at least about 400 consecutive amino acids, at least about 500 consecutive amino acids, at least about 600 consecutive amino acids, at least about 700 consecutive amino acids, or at least about 800 consecutive amino acids of SEQ ID NOS: 79, 88, 150, 158- 160, or 161. Fragments include all possible amino acid lengths between about 8 and 100 about amino acids, for example, lengths between aboui 10 and aboui 100 amino acids, between about 15 and about 100 amino acids, between about 20 and about 100 amino acids, between about 35 and about 100 amino acids, between about 40 and about 100 amino acids, between about 50 and about 100 amino acids, between about 70 and about 100 amino acids, between aboui 75 and aboui 100 amino acids, or between about 80 and about 100 amino acids. These peptide fragments can be obtained commercially or synthesized via liquid phase or solid phase synthesis methods (Atherton et al., ( 1989) Solid Phase Peptide Synthesis: a Practical Approach. IRL Press, Oxford, England), The FGFR fusion peptide fragments can be isolated from a natural source, genetically engineered, or chemically prepared. These methods are well known in the art,
[80185] A FGFR fusion molecule inhibitor can be a protein, such as an antibody
(monoclonal, polyclonal, humanized, chimeric, or fully human), or a binding fragment thereof, directed against a FGFR fusion moleculeof the invention. An antibody fragment can be a form of an antibody other than the full-length form and includes portions or components that exist within foil-length antibodies, in addition to antibody fragments that have been engineered. Antibody fragments can include, but are not limited to, single chain Fv (scFv), diabodies, Fv, and (Fab') ?, triabodies, Fc, Fab, CDRL CDR2, CDR3, combinations of CDR's, variable regions, tctrabodies, bifunctionaJ hybrid antibodies, framework regions, constant regions, and the like (see, Maynard et al., (2000) Ann. Rev. Biomed. Eng. 2:339-76; Hudson (1998) Curr. Opi.n. Biotechnol. 9:395-402). Antibodies can be obtained
commercially, custom generated, or synthesized against an antigen of interest according to methods established in the art (see United States Patent Nos. 6,914,128, 5,780,597,
5,81 1 ,523; Roland E, Kontermann and Stefan Dubel (editors), Antibody Engineering, Vol. 1 & Π. (2010) 2nd ed., Springer; Antony S. Dimitrov (editor), Therapeutic Antibodies: Methods and Protocols (Methods in Molecular Biology). (2009), Humana Press; Benny Lo (editor) Antibody Engineering: Methods and Protocols (Methods in Molecular Biology), (2004) Humana Press, each of which are hereby incorporated by reference in their entireties). For example, antibodies directed to a FGFR fusion molecule can be obtained commercially from Abeam, Santa Cruz Biotechnology, Abgent, R&D Systems, Novus Biologicals, etc. Human antibodies directed to a FGFR fusion molecule (such as monoclonal, humanized, fully human, or chimeric antibodies ) can be useful antibody therapeutics for use in humans. In one embodiment, an antibody or binding fragment thereof is directed against SEQ ID NOS: 79, 88, 150, 158-160, or 161 .
[Θ0186] Inhibition of RNA encoding a FGFR fusion molecule can effectively modulate the expression of a FGFR fusion molecule. Inhibitors are selected from fee group comprising: siRNA; interfering RNA or RNAi; dsRNA; RNA Polymerase III transcribed DNAs;
ribozymes; and antisense nucleic acids, which can be RN A, DNA, or an artificial nucleic acid.
[00187] Antisense oligonucleotides, including antisense DNA, RN A, and DN A/RNA molecules, act to directly block the translation of mR A by binding to targeted mRNA and preventing protein translation. For example, antisense oligonucleotides of at least about 15 bases and complementary to unique regions of the DNA sequence encoding a FGFR fusion molecule can be synthesized, e.g., by conventional phosphodiester techniques (Dallas et al., (2006) Med. Set «#.12(4):RA67-74; alota et al., (2006) Handb. Exp. Pharmacol 173: 173-96; Lutzelburger et al., (2006) Handb, Exp. Pharmacol. 173:243-59). Antisense nucleotide sequences include, but are not limited to: morpholinos, 2'-G-methyl
polynucleotides, DNA, RNA and the like.
[00188] siRNA comprises a double stranded structure containing from about 15 to about 50 base pairs, for example from about 21 to about 25 base pairs, and having a nucleotide sequence identical or nearly identical to an expressed target gene or RNA within the cell. The siRNA comprise a sense RNA strand and a complementary antisense RNA strand annealed together by standard Watson-Crick base-pairing interactions. The sense strand comprises a nucleic acid sequence which is substantially identical to a nucleic acid sequence contained within the target miRNA molecule. "Substantially identical" to a target sequence contained within the target mRNA refers to a nucleic acid sequence that differs from the target sequence by about 3% or less. The sense and antisense strands of the siRNA can comprise two complementary, single- stranded RNA molecules, or can comprise single molecule in which two complementary portions are base-paired and are covalently linked by a single-stranded "hairpin" area. See also, McMnaus and Sharp (2002) Nat Rev Genetics, 3:737-47, and Sen and Blau (2006) FASEB J., 20: 1293-99, the entire disclosures of which are herein incorporated by reference. [0(5189] The siRNA can be altered RNA that differs from naturally-occurring RNA by the addition, deletion, substitution and/or alteration of one or more nucleotides. Such alterations can include addition of non-nucleotide material, such as to the end(s) of the siRNA. or to one or more internal nucleotides of the siRNA, or modifications that make the siRNA resistant to nuclease digestion, or the substitution of one or more nucleotides in the siRNA with deoxyribo-nucleotides. One or both strands of the siRNA can also comprise a 3' overhang. As used herein, a 3' overhang refers to at least one unpaired nucleotide extending from the 3'- end of a duplexed RNA strand. For example, the siRNA can comprise at least one 3' o verhang of from 1 to about 6 nucleotides (which includes ribonucleotides or
dcoxyribonucleotides) in length, or from 1 to about 5 nucleotides in length, or fro 1 to about 4 nucleotides in length, or from about 2 to about 4 nucleotides in length. For example, each strand of the siRNA can comprise 3' overhangs of dithymid lic acid ('ΤΓ') or diuridylic acid ("uu").
[00190] siRNA can be produced chemically or biologically, or can be expressed from a recombinant plasmid or viral vector (for example, see U.S. Patent No. 7,294,504 and U.S. Patent No. 7,422,896, the entire disclosures of which are herein incorporated by reference). Exemplary methods for producing and testing dsRNA or siRNA molecules are described in U.S. Patent Application Publication No. 2002/0173478 io Gewirtz, U.S. Patent No. 8,071,559 to Harmon ct al, and in U.S. Patent No. 7,148,342 to Tolentino et ai, the entire disclosures of which are herein incorporated by reference.
[00191] In one embodiment, an siRNA directed to a human nucleic acid sequence comprising a FGFR fusion molecule can be generated against any one of SEQ ID NOS: 80- 82, 84, 94-144, or 145. In another embodiment, an siRNA directed io a human nucleic acid sequence comprising a breakpoint of anJFGFR fusion molecule can be generated against any one of SEQ ID NOS: 1 -77, 80-82, 84- 144, or 145, In one embodiment, the hairpin sequences targeting the FGFR3 gene comprise SEQ ID NOS: 183, 184, or 185.
[Θ0192] RNA polymerase III transcribed DNAs contain promoters, such as the U6 promoter. These DNAs can be transcribed to produce small hairpin RNAs in the cell that can function as siRNA or linear RNAs, which can function as antisense RNA. The FGFR fusio molecule inhibitor can comprise ribonucleotides, deoxyribonucleotides, synthetic nucleotides, or any suitable combination such that the target RNA and/or gene is inhibited. In addition, these forms of nucleic acid can be single, double, triple, or quadruple stranded, (see for example Bass (2001) Nature, 411 :428-429; Elbashir ei al, (2001 ) Nature, 411 :494 498; U.S. Patent No. 6,509, 154; U.S. Patent Application Publication No. 2003/0027783; and PCT Publication Nos. WO 00/044895, WO 99/032619, WO 00/01846, WO 01/029058, WO 00/044914).
[00193] FOFR fusion molecule inhibitor can be a small molecule that binds to a FOFR fusion protein described herein and disrupts its function. Small molecules are a diverse group of synthetic and natural substances generally having low molecular weights. They can be isolated from natural sources (for example, plants, fungi, microbes and the like), are obtained commercially and/or available as libraries or collections, or synthesized. Candidate small molecules that inhibit a FGFR fusion protein can be identified via in silica screening or high- through-put (HTP) screening of combinatorial libraries according to methods established in the art (e.g., see Poryrailo et al., (2011) ACS Comb Sci, 13(6):579-633; Mensch et al., (2009) JPharm Sci, 98(12):4429-68; Schnur (2008) Curr Opin Drug Discov Devel. l l(3):375-80; and Jhoti (2007) Ernst Schering Found Synip Proc. (3): 169-85, each of which are hereby incorporated by reference in their entireties.) Most conventional pharmaceuticals, such as aspirin, penicillin, and many chemotherapeutics, are small molecules, can be obtained commercially, can be chemically synthesized, or can be obtained from random or combinatorial libraries as described below (see, e.g., Werner ei al., (2006) Brief Fund.
Genomic Proteomic 5(1 ): 32-6).
[ΘΘΙ94] Non-limiting examples of FGFR fusion molecule inhibitors include the FGFR inhibitors AZD4547 (see Gavine et al., (2012) Cancer Res, 72(8); 2045-56; see also PCT Application Publication No. WO 2008/075068, each of which are hereby incorporated by reference in their entireties); NVP-BGJ398 (see Guagna.no et al., (2011) J. Med, Chem., 54:7066-7083; see also U.S. Patent Application Publication No. 2008-0312248 Al, each of which are hereby incorporated by reference in their entireties); PD 173074 (see Guagnano et al, (201 1 ) /. Med. Chem., 54:7066-7083; see also Mohammad! et al., ( 1998) EMBO J., 17:5896-5904, each of which are hereby incorporated by reference in their entireties); NF449 (HMD Millipore (Billerica, MA) Cat. No. 480420; see also Krejci, (201 ) the Journal of Biological Chemistry!, 285(27):20644~20653, which is hereby incorporated by reference in its entirety): LY2874455 (Active Biochem; see Zhao et al. (201 1) Mo I Cancer Then (1 1):2200- 10; see also PCT Application Publication No. WO 2010129509, each of which are hereby incorporated by reference in their entireties); TKT258 (Dovitinib); BJBF- 1 120 (mtedam'b- Vargatei); BMS-582664 (Brivanib aiamnate); AZD-2171 (Cediranib); TSU-68 (Orantinib); AB- 101Q (Masitinib); AP-24534 (Ponatinib); and E-7080 (by Eisai). A non- limiting example of an FGFR fission molecule inhibitor includes the TACC inhibitor KHS101 ( Wurdak et al., (2010) PNAS, 107(38): 16542-47, which is hereby incorporated by reference in its entirety).
[001.95] Structures of FGFR fusion molecule inhibitors useful for the invention; include, but are not limited to: the FGFR inhibitor AZD4547,
; the FGFR inhibitor NVP-BGJ398,
the FGFR inhibitor PD 173074,
; the FGFR inhibitor LY2874455
; and the FGFR inhibitor NF449 (EMT3 Millipore (Billerica,
MA) Cat. No. 480420),
00196] Other FGFR inhibitors include, but are not limited to:
BMS-582664 AZD-2171
f anib alsn:n3te Cediranib
•sto!- yers Squibb) (AstfsZeneca)
•V!asitifiib
AB Science)
(Ariad Pharmaceuticals) , an 90i97] A structure of an FGFR fusion molecule inhibiior useful for the invention include. but is not limited to the TACC inhibitor KHS 101,
Assessment and Therapuetic Treatment
[©0198] The invention provides a method of decreasing the growth of a solid tumor in a subject. The tumor is associated with, but not limited to, glioblastoma multiforme, breast cancer, lung cancer, prostate cancer, or colorectal carcinoma. In one embodiment, the method comprises detecting the presence of a FGFR fusion molecule in a sample obtained from a subject. In some embodiments, the sample is incubated with an agent that binds to an FGFR fusion molecule, such as an antibody, a probe, a nucleic acid primer, and the like. In further embodiments, the method comprises administering to the subject an effective amount of a FGFR fusion molecule inhibitor, wherein the inhibitor decreases the size of the solid tumor.
[0019.9] The invention also provides a method for treating or preventing a gene-fusion associated cancer in a subject. In one embodiment, the gene-fusion associated cancer comprises an epithelial cancer. In one embodiment, the gene-fusion associated cancer comprises glioblastoma multiforme, breast cancer, lung cancer, prostate cancer, or colorectal carcinoma, in some embodiments, the epithelial cancer comprises bladder urothelial carcinoma, breast carcinoma, colorectal cancer, prostate carcinoma, lung squamous cell carcinoma, head and neck squamous ceil carcinoma, or a combination of the epithelial cancers decribed, in one embodiment, the method comprises detecting the presence of a FGFR fitsion molecule in a sample obtained from a subject, the presence of the fusion being indicative of a gene-fusion associated cancer, and, administering to the subject in need a therapeutic treatment against a gene-fusion associated cancer. In some embodiments, the sample is incubated with an agent that binds to an FGFR fusion molecule, such as an antibody, a probe, a nucleic acid primer, and the like,
[ΘΘ2 0] The invention also provides a method for decreasing in a subject in need thereof the expression level or activity of a fusion protein comprising the tyrosine kinase domain of an FGFR protein fused to a polypeptide that eonstitutively activates the tyrosine kinase domain of the FGFR protein. In some embodiments, the method comprises obtaining a biological sample from the subject. In some embodiments, the sample is incubated with an agent that binds to an FGFR fusion molecule, such as an antibody, a probe, a nucleic acid primer, and the like. In some embodiments, the method comprises administering to the subject a therapeutic amount of a composition comprising an admixture of a pharmaceutically accepta ble carrier an inhibitor of the fusion protein of the invention. In another embodiment, the method further comprises determining the fusion protein expression level or activity. In another embodiment, the method further comprises detecting whether the fusion protein expression level or activity is decreased as compared to the fusion protein expression level or activity prior to administration of the composition, thereby decreasing the expression level or activity of the fission protein. In some embodiments, the fission protein is an FGFR-TACC fusion protein.
[0(52111] The administering step in each of the claimed methods can comprise a drug administration, such as FGFR fusion molecule inhibitor (for example, a pharmaceutical composition comprising an antibody that specifically binds to a FGFR fusion molecule or a fragment thereof; a small molecule that specifically binds to a FGFR protein; a small molecule that specifically binds to a TACC protein; an antisense RNA or antisense DNA that decreases expression of a FGFR fusion molecule; a siRNA that specifically targets a gene encoding a FGFR fusion molecule; or a combination thereof). In one embodiment, the therapeutic molecule to be administered comprises a polypeptide of a FGFR fusion molecule, comprising at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 93%, at least about 95%, at least about 97%, at least about 98%, at least about 99%, or 100% of the amino acid sequence of SEQ ID NOS: 79, 88, 150, 158-160, or 161, and exhibits the function of decreasing expression of such a protein, thus treating a gene fusion- associated cancer. In another embodiment, administration of the therapeutic molecule decreases the size of the solid tumor associated with glioblastoma multiforme, breast cancer, lung cancer, prostate cancer, or colorectal carcinoma.
[Θ02Θ2] In another embodiment, the therapeutic molecule to be administered comprises an siRNA directed to a human nucleic acid sequence comprising a FGFR fusion molecule. In one embodiment, the siRNA. is directed to any one of SEQ ID NOS: 80-82, 84, 94-144, or 145. In another embodiment, the siRNA is directed to any one of SEQ ID NOS: 1-77, 80-82, 84- 144, or 145. In a further embodiment, the therapeutic molecule to be administered comprises an antibody or binding fragment thereof, which is directed against SEQ ID NOS: 79, 88, 150, 158- 160, or 161. In some embodiments, the therapeutic molecule to be administered comprises a small molecule that specifically binds to a FOFR protein, such as AZD4547, VP-BGJ398, PD 173074, NF449, TK1258, BIBF-1120, BMS-582664, AZD- 2171, TSU68, AB 1010, AP24534, E-7080, or LY2874455. In other embodiments, the therapeutic molecule to be administered comprises a small molecule that specifically binds to a TACC protein, such as KFIS IOI .
[Θ02Θ3] Irs one embodiment, the invention provides for the detection of a chromosomal rearrangement at given chromosomal coordinates. In another embodiment, the detection or determination comprises nucleic acid sequencing, selective hybridization, selective amplification, gene expression analysis, or a combination thereof. In another embodiment, the detection or determination comprises protein expression analysis, for example by western blot analysis, ELISA, or other antibody detection methods.
[Θ02Θ4] In one embodiment, the biological sample comprises neuronal ceils, serum, bone marrow, blood, peripheral blood, lymph nodes, cerebro- spinal fluid, urine, a saliva sample, a buccal swab, a serum sample, a sputum sample, a lacrimal secretion sample, a semen sample, a vaginal secretion sample, a fetal tissue sample, or a combination thereof.
[00205] A FOFR fusion molecule, for example, a fusion between FGFR1 , FGFR2, FGFR3, or any other FGFR, and TACCl, TACC2, TACC3 or any other TACC, can be determined at the level of the DNA, RNA, or polypeptide. Optionally, detection can be determined by performing an oligonucleotide ligation assay, a confirmation based assay, a hybridization assay, a sequencing assay, an allele-specific amplification assay, a
microsequencing assay, a melting curve analysis, a denaturing high performance liquid chromatography (DHPLC) assay (for example, see Jones et ai, (2000) Hum Genet., i06(6):663-8), or a combination thereof. In one embodiment, the detection is performed by sequencing all or part of a FGFR fusion molecule ( e.g., a FGFR1 -TACCl , FGFR2-TACC2, FGFR3-TACC3 or other FGFR-TACC nucleic acid, or a FGFR1 , TACCl, FGFR2, TACC2, FGFR3, TACC3 or other FGFR or TACC nucleic acid), or by selective hybridization or amplification of all or part of a FGF fusion molecule (e.g., a FGFR 1 -TACCl , FOFR2- TACC2, FGFR3-TACC3 or other FGFR-TACC nucleic acid, or a FGFR1, TACCl, FGFR2, TACC2, FGFR3, TACC3 or other FGFR or TACC nucleic acid). A FGFR fusion molecule specific amplification (e.g., a FGFRl-TACCl, FGFR2-TACC2, FGFR3 -TACC3 or other FGFR-TACC nucleic acid specific amplification) can be carried out before (he fusion identification step.
[§(1206] The invention provides for a method of detecting a chromosomal alteration in a subject afflicted with a gene-fusion associated cancer. In one embodiment, the chromosomal alteration is an in-frame fused transcript described herein, for example an FGFR fusion molecule. In some embodiments, the chromosomal alteration is a chromosomal
translocation, for example an FGFR fusion molecule. An alteration in a chromosome region occupied by a FGFR fusion molecule, such as a FGFRl-TACCl, FGFR2-TACC2, FGFR3- TACC3 or other FGFR-TACC nucleic acid, can be any form of mutation(s), deietion(s), rearrangement(s) and/or insertions in the coding and/or non-coding region of the locus, alone or in various combination^). Mutations can include point mutations. Insertions can encompass the addition of one or several residues in a coding or non-coding portion of the gene locus. Insertions can comprise an addition of between 1 and 50 base pairs in the gene locus. Deletions can encompass any region of one, two or more residues in a coding or non- coding portion of the gene locus, such as from two residues up to the entire gene or locus. Deletions can affect smaller regions, such as domains (introns) or repeated sequences or fragments of less than about 50 consecutiv e base pairs, although larger deletions can occur as well. Rearrangement includes inversion of sequences. The alteration in a chromosome region occupied by a FGFR fusion molecule, e.g., a FGFRl -TACCl , FGFR2-TACC2, FGFR3-TACC3 or other FGFR-TACC nucleic acid, can result in amino acid substitutions, RNA splicing or processing, product instability, the creation of stop eodons, production of oncogenic fusion proteins, frame-shift mutations, and/or truncated polypeptide production. The alteration can result in the production of a FGFR fusion molecule, for example, one encoded by a FGFRl-TACCl, FGFR2-TACC2, FGFR3-TACC3 or other FGFR-TACC nucleic acid, with altered function, stability, targeting or structure. The alteration can also cause a reduction, or even an increase in protein expression. In one embodiment, the alteration in the chromosome region occupied by a FGFR fusion molecule can comprise a chromosomal rearrangement resulting in the production of a FGFR fusion molecule, such as a FGFRl -TACC l , FGFR2-TACC2, FGFR3-TACC3 or other FGFR-TACC fusion. This alteration can be determined at the level of the DNA, RNA, or polypeptide. In another embodiment, the detection or determination comprises nucleic acid sequencing, selective hybridization, selective amplification, gene expression analysis, or a combination thereof. In another embodiment, the detection or determination comprises protein expression analysis, for example by western blot analysis, ELISA, or other antibody detection methods. In one embodiment, the coordinates comprising FGFR1 translocations comprise chr8:38,268,656- 38,325,363. In another embodiment, the coordinates comprising FGFR2 translocations comprise chrl0:123,237,844-123,357,972. In a further embodiment, the coordinates comprising FGFR3 translocations comprise chr4:l ,795,039- 1,810,599. In yet another embodiment, the coordinates comprising FGFR4 translocations comprise chr5: 176,513,921- 176,525,126. In one embodiment, the coordinates comprising TACC1 translocations comprise ehr8:38,644,722-38, 710,546. In another embodiment, the coordinates comprising TACC2 translocations comprise chrl 0:123,748,689-124,014,057. In a farther embodiment, the coordinates comprising TACC3 translocations comprise chr4: 1 ,723,217- 1 ,746,905.
[00207] The present invention provides a method for treating a gene-fusion associated cancer in a subject in need thereof. In one embodiment, the method comprises obtaining a sample from the subject to determine the le vel of expression of an FGFR fusion molecule in the subject. In some embodiments, the sample is incubated with an agent that binds to an FGFR fusion molecule, such as an antibody, a probe, a nucleic acid primer, and the like. In another embodiment, the detection or determination comprises nucleic acid sequencing, selective hybridization, selective amplification, gene expression analysis, or a combination thereof. In another embodiment, the detection or determination comprises protein expression analysis, for example by western blot analysis, ELISA, or other antibody detection methods. In some embodiments, the method further comprises assessing whether to administer a FGFR fusion molecule inhibitor based on the expression pattern of the subject. In farther embodiments, the method comprises administering a FGFR fusion molecule inhibitor to the subject. In one embodiment, the gene-fusion associated cancer comprises an epithelial cancer. In one embodiment, the gene-fusion associated cancer comprises glioblastoma multiforme, breast cancer, lung cancer, prostate cancer, or colorectal carcinoma. In some embodiments, the epithelial cancer comprises bladder urothelial carcinoma, breast carcinoma, colorectal cancer, prostate carcinoma, lung squamous ceil carcinoma, head and neck squamous ceil carcinoma, or a combination of the epithelial cancers decribed.
[Θ82Θ8] In one embodiment, the invention provides for a method of detecting the presence of altered RNA expression of an FGFR fusion molecule in a subject, for example one afflicted with a gene-fusion associated cancer. In another embodiment, the invention provides for a method of detecting the presence of an FGFR fusion molecule in a subject. In some embodiments, the method comprises obtaining a sample from the subject to determine whether the subject expresses an FGFR fusion molecule, in some embodiments, the sample is incubated with an agent that binds to an FGFR fusion molecule, such as an antibody, a probe, a nucleic acid primer, and the like. In other embodiments, the detection or determination comprises nucleic acid sequencing, selective hybridization, selective amplification, gene expression analysis, or a combination thereof. In another embodiment, the detection or determination comprises protein expression analysis, for example by western blot analysis, ELISA, or other antibody detection methods. In some embodiments, the method further comprises assessing whether to administer a FGFR fission molecule inhibitor based on the expression pattern of the subject. In further embodiments, the method comprises administering a FGFR fusion molecule inhibitor to the subject. Altered RNA expression includes the presence of an altered RNA sequence, the presence of an altered RN A splicing or processing, or the presence of an altered quantity of RN A. These can be detected by various techniques known in the art, including sequencing all or part of the RNA or by selecti ve hybridization or selective amplification of all or part of the RNA. In a further embodiment, the method can comprise detecting the presence or expression of a FGFR fusion molecuie, such as one encoded by a FGFR l -TACC1 , FGFR2-TACC2, FGFR3-TACC3 or other FGFR-TACC nucleic acid. Altered polypeptide expression includes the presence of an altered polypeptide sequence, the presence of an altered quantity of polypeptide, or the presence of an altered tissue distribution. These can be detected by various techniques known in the art, including by sequencing and/or binding to specific ligands (such as antibodies ). In one embodiment, the detecting comprises using a northern blot; real time PGR and primers directed to SEQ ID NOS: 80-82, 84, 94- 144, or 145; a ribonuclease protection assay; a hybridization, amplification, or sequencing technique to detect an FGFR fusion molecule, such as one comprising SEQ ID NOS: 80-82, 84, 94- 144, or 145: or a combination thereof. In another embodiment, the PGR primers comprise SEQ ID NOS: 162, 163, 164, or 165. In a further embodiment, primers used for the screening of FGFR fusion molecules, such as FGFR-TACC fusions, comprise SEQ ID NOS: 166, 167, 168, or 169. In some embodiments, primers used for genomic detection of an FGFR3-TACC3 fusion comprise SEQ ID NOS: 170 and 171.
[1)8209] Various techniques known in the art can be used to detect or quantify altered gene or RNA expression or nucleic acid sequences, which include, but are not limited to, hybridization, sequencing, amplification, and/or binding to specific iigands (such as antibodies). Other suitable methods include allele-speciiic oligonucleotide (ASO), oligonucleotide ligation, allele-specific amplification, Southern blot (for DNAs), Northern blot (for RIM As), single-stranded conformation analysis (SSC A), PFGE, fluorescent in situ hybridization (FISH), gel migration, clamped denaturing gel electrophoresis, denaturing HLPC, melting curve analysis, heteroduplex analysis, RNase protection, chemical or enzymatic mismatch cleavage, ELISA, radio-imniunoassays (RIA) and imrauno-enzymatic assays (IEMA).
[00210] Some of these approaches (such as SSCA and constant gradient gel
electrophoresis (CGGE)) are based on a change in electrophoretic mobility of the nucleic acids, as a result of the presence of an altered sequence. According to these techniques, the altered sequence is visualized by a shift in mobility on gels. The fragments can then be sequenced to confirm the alteration. Some other approaches are based on specific hybridization between nucleic acids from the subject and a probe specific for wild type or altered gene or RNA. T e probe can be in suspension or immobilized on a substrate. The probe can be labeled to facilitate detection of hybrids. Some of these approaches are suited for assessing a polypeptide sequence or expression level, such as Northern blot, ELISA and RIA. These latter require the use of a ligand specific for the polypeptide, for example, the use of a specific antibody.
[00211] Hybridization. Hybridization detection methods are based on the formation of specific hybrids between complementary nucleic acid sequences that serve to detect nucleic acid sequence alterations). A detection technique involves the use of a nucleic acid probe specific for a wild type or altered gene or RNA, followed by ihe detection of ihe presence of a hybrid. The probe can be in suspension or immobilized on a substrate or support (for example, as in nucleic acid array or chips technologies). The probe can be labeled to facilitate detection of hybrids. In one embodiment, the probe according to the invention can comprise a nucleic acid directed to SEQ ID NOS: 80-82, 84, 94-144, or 145. For example, a sample from the subject can be contacted with a nucleic acid probe specific for a gene encoding a FGFR fusion molecule, and the formation of a hybrid can be subsequently assessed. In one embodiment, the method comprises contacting simultaneously the sample with a set of probes that are specific for an FGFR fusion molecule. Also, various samples from various subjects can be investigated in parallel. [0(5212] According to the invention, a probe can be a polynucleotide sequence which is complementary to and specifically hybridizes with a, or a target portion of a, gene or RNA corresponding to a FGF fusion molecule. Useful probes are those that are complementary to the gene, RNA, or target portion thereof. Probes can comprise single-stranded nucleic acids of between 8 to 1000 nucleotides in length, for instance between 10 and 800, between 15 and 700, or between 20 and 500. Longer probes can be used as well. A useful probe of the invention is a single stranded nucleic acid molecule of between 8 to 500 nucleotides in length, which can specifically hybridize to a region of a gene or RNA that corresponds to a FGFR fusion molecule.
[00213] The sequence of the probes can be derived from the sequences of the FGFR fusion genes provided herein. Nucleotide substitutions can be performed, as well as chemical modifications of the probe. Such chemical modifications can be accomplished to increase the stability of hybrids (e.g., intercalating groups) or to label the probe. Some examples of labels include, without limitation, radioactivity, fluorescence, luminescence, and enzymatic labeling.
[00214] A guide to the hybridization of nucleic acids is found in e.g., Sambrook, ed., Molecular Cloning: A Laboratory Manual (3id Ed.), Vols. 1-3, Cold Spring Harbor
Laboratory, 1989; Current Protocols In Molecular Biology, Ausubel, ed. John Wiley & Sons, Inc., New York, 2001 ; Laboratory Techniques In Biochemistry And Molecular Biology: ybridjzation
Tijssen, ed. Elsevier, N.Y., 1993.
[00215] Sgq jencing. Sequencing can be carried out using techniques well known in the art, using automatic sequencers. The sequencing can be performed on the complete FGFR fusion molecule or on specific domains thereof.
[00216] Amplification. Amplification is based on the formation of specific hybrids between complementary nucleic acid sequences that serve to initiate nucleic acid reproduction. Amplification can be performed according to various techniques known in the art, such as by polymerase chain reaction (PGR), ligase chain reaction (LCR), strand displacement amplification (SDA) and nucleic acid sequence based amplification (NASBA). These techniques can be performed using commercially available reagents and protocols. Useful techniques in the art encompass real-time PGR, allele-specific PCR, or PCR based single-strand conformational polymorphism (SSCP). Amplification usually requires the use of specific nucleic acid primers, to initiate the reaction. For example, nucleic acid primers useful for amplifying sequences corresponding to a FGFR fusion molecule are able to specifically hybridize with a portion of the gene locus that flanks a target region of the locus. In one embodiment, amplification comprises using forward and reverse PGR primers directed to SEQ ID NOS: 80-82, 84, 94- 144, or 145. Nucleic acid primers useful for amplifying sequences from a FGFR fusion molecule (e.g., a FGFRl-TACCl, FGFR2-TACC2, FGFR3- TACC3 or other FGFR-TACC nucleic acid); the primers specifically hybridize with a portion of an FGFR fusion molecule. In certain subjects, the presence of an FGFR fusion molecule corresponds to a subject with a gene fusion-associated cancer. In one embodiment, amplification can comprise using forward and reverse PGR primers comprising nucleotide sequences of SEQ ID NOS: 80-82, 84, 94- 144, or 145.
[0(5217] Non-limiting amplification methods include, e.g., polymerase chain reaction, PCR (PGR Protocols, A Guide To Methods And Applications, ed. lnnis, Academic Press, N.Y., 1990 and PCR Strategies. 1995, ed. Innis, Academic Press, Inc., N.Y.); ligasc chain reaction (LCR) (Wu (1989) Genomics 4:560; Landegren ( 1988) Science 241 : 1077; Barringer (1990) Gene 89: 1 17); transcription amplification (Kwoh ( 1989) PNAS 86: 1 173); and, self-sustained sequence replication (Guatelli (1990) PNAS 87: 1874); Q Beta replicase amplification (Smith (1997) J. Clin. Microbiol. 35: 1477-1491), automated Q-beta replicase amplification assay (Burg (1996) Mol. Cell Probes: 10:257-271) and other RNA polymerase mediated techniqites (e.g., NASBA, Cangene, Mississauga, Ontario; see also Berger (1987) Methods Enzymol. 152:307-316; U.S. Pat Nos. 4,683,195 and 4,683,202; and Sooknanan (3995) Biotechnology 13:563-564). All the references stated above are incorporated by reference in their entireties.
[00218] The invention provides for a nucleic acid primer, wherein the primer can be complementary to and hybridize specifically to a portion of a FGFR fusion molecule, such as a FGFRl-TACCl, FGFR2-TACC2, FGFR3-TACC3 or other FGFR-TACC nucleic acid (e.g., DNA or RNA) in certain subjects having a gene fusion-associated cancer. In one embodiment, the gene-fusion associated cancer comprises glioblastoma multiforme, breast cancer, lung cancer, prostate c ancer, or colorectal carcinoma. Primers of the invention can be specific for fusion sequences in a FGFRl-TACC l, FGFR2-TACC2, FGFR3-TACC3 or other FGFR-TACC nucleic acid (DNA or R A). By using such primers, the detection of an amplification product indicates the presence of a fusion of a FGFR1 and TACC1 , FGFR2 and TACC2, FGFR3 and TACC3 or other FGFR and TACC nucleic acid. Examples of primers of this invention can be single-stranded nucleic acid molecules of about 5 to 60 nucleotides in length, or about 8 to about 25 nucleotides in length. The sequence can be derived directly from the sequence of a FGFR fusion molecule, e.g. FGFR 1 -TACC I, FGFR2-TACC2, FGFR3-TACC3 or other FGFR-TACC nucleic acid. Perfect complementarity is useful to ensure high specificity; however, certain mismatch can be tolerated. For example, a nucleic acid primer or a pair of nucleic acid primers as described above can be used in a method for detecting the presence of a gene fission-associated cancer in a subject. In one embodiment, primers can be used to detect an FGFR fusion molecule, such as a primer comprising SEQ ID OS: 80-82, 84, 94-144, or 145; or a combination thereof. In another embodiment, the PGR primers comprise SEQ ID NOS: 162, 163, 164, or 165. In a further embodiment, primers used for the screening of FGFR fusion molecules, such as FGFR-TACC fusions, comprise SEQ ID NOS: 166, 167, 168, or 169. In some embodiments, primers used for genomic detection of an FGFR3-TACC3 fusion comprise SEQ ID NOS: 170 and 171.
[00219] Specific Ligand Binding. As discussed herein, a nucleic acid encoding a FGFR fusion molecule or expression of a FGF fusion molecule, can also be detected by screening for alteration(s) in a sequence or expression level of a polypeptide encoded by the same. Different types of ligands can be used, such as specific antibodies. In one embodiment, the sample is contacted with an antibody specific for a polypeptide encoded by a FGFR fusion molecule and the formation of an immune complex is subsequently determined. Various methods for detecting an immune complex can be used, such as ELISA, radioimmunoassays (RIA) and immuno-enzymatic assays (IEMA).
[0Θ22Θ] For example, an antibody can be a polyclonal antibody, a monoclonal antibody, as well as fragments or derivatives thereof having substantially the same antigen specificity. Fragments include Fab, Fab'2, or CDR regions. Derivatives include single-chain antibodies, humanized antibodies, or poly-functional antibodies. An antibody specific for a polypeptide encoded by a FGFR fusion molecule can be an antibody that selectively binds such a polypeptide. In one embodiment, the antibody is raised against a polypeptide encoded by a FGFR fusion molecule ( such as FGFRl -TACCl , FGFR2-TACC2, FGFR3-TACC3 or other FGFR-TACC fusion) or an epitope-containing fragment thereof. Although non-specific binding towards other antigens can occur, binding to the target polypeptide occurs with a higher affinity and can be reliably discriminated from non-specific binding. In one embodiment, the method can comprise contacting a sample from the subject with an antibody specific for a FGFR fusion molecule, and determining the presence of an immune complex. Optionally, the sample can be contacted to a support coated with antibody specific for a FGFR fusion molecule. In one embodiment, the sample can be contacted simultaneously, or in parallel, or sequentially, with various antibodies specific for different forms of a FGFR fusion molecule, e.g., FGFR1 -TACC1 , FGFR2-TACC2, FGFR3-TACC3 or other FGFR- TACC fusion.
[ΘΘ221 ] The invention also provides for a diagnostic kit comprising products and reagents for detecting in a sample from a subject the presence of a FGFR fusion molecule. The kit can be useful for determining whether a sample from a subject exhibits increased or reduced expression of a FGFR fusion molecule. For example, the diagnostic kit according to the present invention comprises any primer, any pair of primers, any nucleic acid probe and/or any ligand, or any antibody directed speci fical ly to a FGFR fusion molecule. The diagnostic kit according to the present invention can further comprise reagents and/or protocols for performing a hybridization, amplification, or antigen-antibody immune reaction. In one embodiment, the kit can comprise nucleic acid primers that specifically hybridize to and can prime a polymerase reaction from a FGFR fusion molecule comprising SEQ ID NOS: 80-82, 84, 94- 144, or 145, or a combination thereof. In one embodiment, primers can be used to detect a FGFR fusion molecule, such as a primer directed to SEQ ID NOS: 80-82, 84, 94- 144, or 145; or a combination thereof. In another embodiment, the PGR primer comprises SEQ ID NOS: 162, 163, 164, 165, 166, 167, 168, or 169. In a further embodiment, primers used for the screening of FGFR fusion molecules, such as FGFR-TACC fusions, comprise SEQ ID NOS: 166, 1 67, 168, or 169. In some embodiments, primers used for genomic detection of an FGFR3-TACC3 fusion comprise SEQ ID NOS: 170 and 171. In some embodiments, the kit comprises an antibody that specifically binds to a FGFR fusion molecule comprising SEQ ID NOS: 79, 85-89, 150, 158- 160, or 161 , wherein the antibody will recognize the protein only when a FGFR fusion molecule is present
{00222] The diagnosis methods can be performed in vitro, ex vivo, or in vivo. These methods utilize a sample from the subject in order to assess the status of a FGFR fission molecule. The sample can be any biological sample derived from a subject, which contains nucleic acids or polypeptides. Examples of such samples include, but are not limited to, fluids, tissues, cell samples, organs, and tissue biopsies. Non-limiting examples of samples include blood, liver, plasma, serum, saliva, urine, or seminal fluid. The sample can be coliecied according to conventional techniques and used directly for diagnosis or stored. The sample can be treated prior to performing the method, in order to render or improve availability of nucleic acids or polypeptides for testing. Treatments include, for instance, lysis (e.g., mechanical, physical, or chemical), centrifugation. The nucleic acids and/or polypeptides can be pre-purified or enriched by conventional techniques, and/or reduced in complexity. Nucleic acids and polypeptides can also be treated with enzymes or other chemical or physical treatments to produce fragments thereof. In one embodiment, the sample is contacted with reagents, such as probes, primers, or ligands, in order to assess the presence of a FGFR fusion molecule. Contacting can be performed in any suitable device, such as a plate, tube, well, or glass, in some embodiments, the contacting is performed on a substrate coated with the reagent, such as a nucleic acid array or a specific ligand array. The substrate can be a solid or semi-solid substrate such as any support comprising glass, plastic, nylon, paper, metal, or polymers. The substrate can be of various forms and sizes, such as a slide, a membrane, a bead, a column, or a gel. The contacting can be made under any condition suitable for a complex to be formed between the reagent and the nucleic acids or polypeptides of the sample.
Nucleic Acid Delivery Methods
[0(5223] Delivery of nucleic acids into v iable cells can be effected ex vi vo, in siiu, or in vivo by use of vectors, such as viral vectors (e.g., ientiviras, adenovirus, adeno-associated vims, or a retrovirus), or ex vivo by use of physical DNA transfer methods (e.g., liposomes or chemical treatments). Non-limiting techniques suitable for the transfer of nucleic acid in to mammalian cells in vitro include the use of liposomes, eiectroporation, microinjection, cell fusion, DEAE-dextran, and the calcium phosphate precipitation method (See, for example, Anderson, Nature, 1998) supplement to 392(6679):25(). introduction of a nucleic acid or a gene encoding a polypeptide of the invention can also be accomplished with
extrachromosomai substrates (transient expression) or artificial chromosomes (stable expression). Cells can also be cultured ex vivo in the presence of therapeutic compositions of the present invention in order to proliferate or to produce a desired effect on or activity in such ceils. Treated cells can then be introduced in vivo for therapeutic purposes. 0Θ224] Nucleic acids can be inserted into vectors and used as gene therapy vectors. A number of viruses have been used as gene transfer vectors, including papo vaviruses, e.g., SV40 (Madzak ei al, ( 1992) J Gen Virol. 73( Ft 6): 1533-6), adenovirus (Berkner ( 1992) Curr Top Microbiol Immunol.158:39-66; Berkner (1988) Biotechniques, 6(7):616-29; Gorziglia and Kapikian ( 1992) J Virol. 66(7):4407-12; Quantin ei al,, ( 1992) Proc Nail Acad Sci U S A. 89(7):2581 -4; Rosenfeld et al., (1992) Cell. 68(1 ): 143-55; Wilkinson et al., (1992) Nucleic Acids Res. 20(9):2233-9; Stratford-Perricaudet et al., (1990) Hum Gene Ther. l (3):241 -56), vaccinia virus (Moss (1992) Curr Opin Bioiechnol. 3(5):518-22), adeno- associated virus (Muzyczka, (1992) Curr Top Microbiol Immunol. 158:97- 129; Ohi et al, (1990) Gene.
89(2):279-82), herpesviruses including HSV and EBV (Margolskee ( 1992) Curr Top Microbiol Immunol. 158:67-95; Johnson et al., (1992) Brain Res Mol Brain ite. l2(i-3):95- 102; Fink et al., (3992) Hum Gene Ther. 3(l):l l-9; Breakefield and Geller (1987) Mol Neurohiol. i(4):339~71 ; Freese et al., (1990) Biochem Pharmacol. 40( 10):2i 89-99), and retroviruses of avian (Bandyopadhy y and Temin ( 1984) Mol Cell Biol. 4(4):749-54;
Petropoulos et al, ( 1992) J Virol. 66(6):3391 -7), murine (Miller et al. ( 1992) Mol Cell Biol. 52(7):3262-72; Miller et al ., (1985) J Virol. 55(3):521-6; Sorge et al, (1984) Mol Cell Biol. 4(9): 1730-7; Mann and Baltimore (1985) J Virol 54(2):401 -7; Miller et al, ( 1988) J Virol. 62(1 1):4337-45), and human origin (Shimada et al., (1991) J Clin invest. 88(3): 1043-7; Helseth et al, (1990) J Virol. 64(12):6314-8; Page et al, (5990) J Virol 64(5 3):5270-6; Buchschacher and Panganiban (1992) J Virol. 66(5):2731-9).
[00225] N on-limiting examples of in vivo gene transfer techniques include transfection with viral (e.g., retroviral) vectors (see U.S. Pat. No. 5,252,479, which is incorporated by reference in its entirety) and viral coat protein- liposome mediated transfection (Dzau et al., (1993) Trends in Biotechnology 1 1 :205-210), incorporated entirely by reference). For example, naked DNA vaccines are generally known in the art; see Brower, (1998) Nature Biotechnology, 16: 1304-1305, which is incorporated by reference in its entirety. Gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (see, e.g., U.S. Pat. No. 5,328,470) or by stereotactic injection (see, e.g., Chen, et al., (1994) Proc. Natl. Acad. Sci. USA 91 :3054-3057). The pharmaceutical preparation of the gene therapy vector can include the gene therapy vector in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded.
Alternatively, where the complete gene delivery vector can be produced intact from recombinant cells, e.g., retroviral vectors, the pharmaceutical preparation can include one or more cells that produce the gene delivery system. [0(5226] For reviews of nucleic acid delivery protocols and methods see Anderson et al. (1992) Science 256:808-813; U.S. Pat Nos. 5,252,479, 5,747,469, 6,017,524, 6,143,290, 6,410,010 6,51 1 ,847; and U.S. Application Publication No. 2002/0077313, which are all hereby incorporated by reference in their entireties. For additional reviews, see Priedmann (1989) Science, 244: 1275-1281; Verma, Scientific American: 68-84 (1990); Miller (1992) Nature, 357: 455-460; Kikuchi et al. (2008) J Dermatol Sci. 50(2):87-98; lsaka et al. (2007) Expert Opin Drug Deliv. 4(5):565 -71; Jager et al.(2007) Curr Gene Ther. 7(4):272-83;
Waehler et al.(2007) Nat Rev Genet. 8(8):573-87; Jensen et al. (2007) Ann Med. 39(2): 108- 15; Herweijer et al. (2007) Gene Ther. 14(2):99- 107; Eliyahu et al. (2005) Molecules 10(l):34-64; and Aitaras et al. (2005) Adv Biochem Eng Biotechnol. 99: 193-260, all of which are hereby incorporated by reference in their entireties.
[00227] A FGFR fusion nucleic acid can also be delivered in a controlled release system. For example, the FGFR fusion molecule can be administered using intravenous infusion, an implantable osmotic pump, a transdermal patch, liposomes, or other modes of admimstration. In one embodiment, a pump can be used (see Sefton (1987) Biomed. Eng. 14:201; Buchwald et al. (1980) Surgery 88:507; Saudek et al. (1989) N. Engl. J. Med. 321 :574). In another embodiment, polymeric materials can be used (see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla. (1974); Controlled Drug Bjogyailaj^ Smolen and Ball (eds.), Wiley, New
York (1984); Ranger and Peppas, ( 1983) /. Macromol. Sci. Rev. Macromol. Chem. 23:61; see also Levy et al (1985) Science 228: 190; During et al. ( 1989) Ann. Neurol. 25:351 ; Howard et al. (1989) J. Neurosurg. 71 : 105). In yet another embodiment, a controlled release system can be placed in proximity of the therapeutic target thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)). Other controlled release systems are discussed in the review by Langer (Science (1990) 249: 1527-1533).
Pharmaceutical Compositions and Administration for Therapy
[Θ0228] An inhibitor of the invention can be incorporated into pharmaceutical compositions suitable for administration, for example the inhibitor and a pharmaceutically acceptable carrier [0(5229] A FGFR fusion molecule or inhibitor of the invention can be administered to the subject once (e.g., as a single injection or deposition). Alternatively, a FGFR fusion molecule or inhibitor can be administered once or twice daily to a subject in need thereof for a period of from about two to about twenty-eight days, or from about seven to about ten days. A FGFR fusion molecule or inhibitor can also be administered once or twice daily to a subject for a period of i , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12 times per year, or a combination thereof. Furthermore, a FGFR fusion molecule or inhibitor of the invention can be co- admini trated with another therapeutic. Where a dosage regimen comprises multiple administrations, the effective amount of the FGFR fusion molecule or inhibitor administered to the subject can comprise the total amount of gene product administered over the entire dosage regimen.
[00230] A FGFR fusion molecule or inhibitor can be administered to a subject by any means suitable for delivering the FGFR fusion molecule or inhibitor to cells of the subject, such as cancer cells, e.g., glioblastoma multiforme, breast cancer, lung cancer, prostate cancer, or colorectal carcinoma. For example, a FGFR. fusion molecule or inhibitor can be administered by methods suitable to transfect cells. Transfection methods for eukatyotic cells are well known in the art, and include direct injection of the nucleic acid into the nucleus or pronucleus of a ceil; electroporation; liposome transfer or transfer mediated by lipophilic materials; receptor mediated nucleic acid delivery, bioballistic or particle acceleration; calcium phosphate precipitation, and transfection mediated by viral vectors.
[00231 j The compositions of this invention can be formulated and administered to reduce the symptoms associated with a gene fusion-associated cancer, e.g., glioblastoma multiforme, breast cancer, lung cancer, prostate cancer, or colorectal carcinoma, by any means that produces contact of the active ingredient with the agent's site of action in the body of a subject, such as a human or animal (e.g., a dog, cat, or horse). They can be administered by any conventional means available for use in conjunction with pharmaceuticals, either as indi vidual therapeutic active ingredients or in a combination of therapeutic active ingredients. They can be administered alone, but are generally administered with a pharmaceutical carrier selec ted on the basis of the chosen route of administration and standard pharmaceutical practice.
[ΘΘ232] A therapeutically effective dose of FGFR fusion molecule or inhibitor can depend upon a number of factors known to those or ordinary skill in the art. The dose(s) of the FGFR fusion molecule inhibitor can vary, for example, depending upon the identity, size, and condition of the subject or sample being treated, further depending upon the route by which the composition is to be administered, if applicable, and the effect which the practitioner desires the a FGFR fusion molecule inhibitor to have upon the nucleic acid or polypeptide of the invention. These amounts can be readily determined by a skilled artisan. Any of the therapeuiic applications described herein can be applied to any subject in need of such therapy, including, for example, a mammal such as a dog, a cat, a cow, a horse, a rabbit, a monkey, a pig, a sheep, a goat, or a human.
[00233] Pharmaceutical compositions for use in accordance with the invention can be formulated in conventional manner using one or more physiologically acceptable carriers or excipienis. The therapeutic compositions of the invention can be formulated for a variety of routes of administration, including systemic and topical or localized administration.
Techniques and formulations generally can be found in Remmington's Pharmaceutical Sciences, Meade Publishing Co., Easton, Pa (2.0"* Ed., 2000), the entire disclosure of which is herein incorporated by reference. For systemic administration, an injection is useful, including intramuscular, intravenous, intraperitoneal, and subcutaneous. For injection, the therapeutic compositions of the invention can be formulated in liquid solutions, for example in physiologically compatible buffers such as Hank's solution or Ringer's solution. In addition, the therapeutic compositions can be formulated in solid form and redissolved or suspended immediately prior to use. Lyophilized forms are also included. Pharmaceutical compositions of the present invention are characterized as being at least sterile and pyrogen- free. These pharmaceutical formulations include formulations for human and veterinary use.
[60234] According to the invention, a pharmaceutically acceptable carrier can comprise any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art. Any conventional media or agent that is compatible with the active compound can be used. Supplementary active compounds can also be incorporated into the compositions.
[00235] A pharmaceutical composition containing FGFR fusion molecule inhibitor can be administered in conjunction with a pharmaceutically acceptable carrier, for any of the therapeutic effects discussed herein. Such pharmaceutical compositions can comprise, for example antibodies directed to a FGFR fusion molecule, or a variant thereof, or antagonists of a FGFR fusion molecule. The compositions can he administered alone or in combination with at least one other agent, such as a stabilizing compound, which can be administered in any sterile, biocompatible pharmaceutical carrier including, but not limited to, saline, buffered saline, dextrose, and water. The compositions can be administered to a patient alone, or in combination with other agents, drugs or hormones.
[ΘΘ236] Sterile injectable solutions can be prepared by incorporating the FGFR fusion molecule inhibitor (e.g., a polypeptide or antibody) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated herein, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated herein. In the case of sterile powders for the preparation of sterile injectable solutions, examples of useful preparation methods are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof,
[Θ0237] In some embodiments, the FGFR fusion molecule inhibitor can be applied via transdermal delivery systems, which slowly releases the active compound for percutaneous absorption. Permeation enhancers can be used to facilitate transdermal penetration of the active factors in the conditioned media. Transdermal patches are described in for example, U.S. Pat. No. 5,407,713; U.S. Pat. No. 5,352,456; U.S. Pat. No. 5,332,213; U.S. Pat. No. 5,336,168; U.S. Pat. No. 5,290,561 ; U.S. Pat. No. 5,254,346; U.S. Pat. No. 5,164,189; U.S. Pat. No. 5,163,899; U.S. Pat. No. 5,088,977; U.S. Pat. No. 5,087,240; U.S. Pat. No.
5,008,1 10; and U.S. Pat. No. 4,921 ,475.
{00238] "Subcutaneous" administration can refer to administration just beneath the skin (i.e., beneath the dennis). Generally, the subcutaneous tissue is a layer of fat and connective tissue that houses larger blood vessels and nerves. The size of this layer varies throughout the body and from person to person. The interface between the subcutaneous and muscle layers can be encompassed by subcutaneous administration. This mode of administration can be feasible where the subcutaneous lay er is sufficiently thin so that the factors present in the compositions can migrate or diffuse from the locus of administration. Thus, where intradermal administration is utilized, the bolus of composition administered is localized proximate to the subcutaneous layer. [00239] Administration of the cell aggregates (such as DP or DS aggregates) is not restricted to a single route, but can encompass administration by multiple routes. For instance, exemplary administrations by multiple routes include, among others, a combination of intradermal and intramuscular administration, or intradermal and subcutaneous administration. Multiple administrations can be sequential or concurrent. Other modes of application by multiple routes will be apparent to the skilled artisan.
[Θ024Θ] In other embodiments, this implantation method will be a one-time treatment for some subjects, in further embodiments of the invention, multiple cell therapy implantations will be required, in some embodiments, the ceils used for implantation will generally be subject-specific genetically engineered cells. In another embodiment, cells obtained from a different species or another individual of the same species can be used. Thus, using such ceils can require administering an immunosuppressant to prevent rejection of the implanted cells. Such methods have also been described in United States Patent No. 7,419,66 land PCT application publication WO 2001/32840, and are hereby incorporated by reference.
[0024 ] A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation or ingestion), transdermal (topical), transmucosal, and rectal administration. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
[Θ0242] Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble ) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringabiiity exists. It must be stable under the conditions of manufacture and storage and must be preserv ed against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, a pharmaceutically acceptable polyol like glycerol, propylene glycol, liquid polyetheylene glycol, and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, ehlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it can be useful to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
[0(5243] Sterile injectable solutions can be prepared by incorporating the inhibitor (e.g., a polypeptide or antibody or small molecule) of the invention in the required amount i an appropriate solvent with one or a combination of ingredients enumerated herein, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated herein. In the case of sterile powders for the preparation of sterile injectable solutions, examples of useful preparation methods are vacuum drying and freeze shying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
[ΘΘ244] Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier and subsequently swallowed.
[60245] Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as macrocrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as aiginic acid, Primogel, or com starch; a lubricant such as magnesium stearate or sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring,
[00246] Systemic administration can also be by transmucosal or transdermal means. For transmucGsal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the foramlation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, deiergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
[0(5247] In some embodiments, the effective amount of the administered FGFR fusion molecule inhibitor is at least about 0.0001 ug/kg body weight, at least about 0.00025 μ^'Ί¾ body weight, at least about 0,0005 ug/kg body weight, at least about 0.00075 ^ig/kg body weight, at least about 0.001 ug/kg body weight, at least about 0.0025 ug/kg body weight, at least about 0.005 ug/kg body weight, at least about 0.0075 ug/kg body weight, at least about 0.01 ug/kg body weight, at least about 0.025 ug/kg body weight, at least about 0.05 ug/kg body weight, at least about 0.075 ug kg body weight, at least about 0.1 ug/kg body weight, at least about 0.25 ug/kg body weight, at least about 0.5 ug/kg body weight, at least about 0.75 ug/kg body weight, at least about 1 ug/kg body weight, at least about 5 ug kg body weight, at least about 10 .ug/kg body weight, at least about 25 ug/kg body weight, at least about 50 ug/kg body weight, at least about 75 μ^Λ^, body weight, at least about 100 ug/kg body weight, at least about 150 ug/kg body weight, at least about 200 ug/kg body weight, at least about 250 ug/kg body weight, at least about 300 ug/kg body weight, at least about 350 ug/kg body weight, at least about 400 g/kg body weight, at least about 450 ug kg body weight, at least about 500 ug/kg body weight, at least about 550 ug/kg body weight, at least about 600 g/kg body weight, at least about 650 ug/kg body weight, at least about 700 ug/kg body weight, at least about 750 μg/kg body weight, at least about 800 ug/kg body weight, at least about 850 μg/ g body weight, at least about 900 ug/kg body weight, at least about 950 ug/kg body weight, at least about 1,000 ug/kg body weight, at least about 2,000 ^ig/kg body weight, at least about 3,000 ug/kg body weight, at least about 4,000 ug/kg body weight, at least about 5,000 ug/kg body weight, at least about 6,000 ^ig/kg body weight, at least about 7,000 tig kg body weight, at least about 8,000 ,u.g/kg body weight, at least about 9,500 pg kg body weight, or at least about 10,000 μg/kg body weight,
{00248] Unless otherwise defined, all technical and scientific tenns used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Exemplary methods and materials are described below, although methods and materials si milar or equivalent to those described herein can also be used in the practice or testing of the present invention.
[00249] All publications and other references mentioned herein are incorporated by reference in their entirety, as if each individual publication or reference were specifically and individually indicated to be incorporated by reference. Publications and references cited herein are not admitted to be prior art.
EXAMPLES
[00250] Examples are provided below to facilitate a more complete understanding of the invention. The following examples illustrate the exemplary modes of making and practicing the invention. However, the scope of the invention is not limited to specific embodiments disclosed in these Examples, which are for purposes of illustration only, since alternative methods can be utilized to obtain similar results.
[00251 j Example 1: Transforming and recurrent fusions of FGFR and TACC gene in glioblastoma
[0(5252] The history of successful targeted therapy of cancer largely coincides with the inactivation of recurrent, oncogenic and addicting gene fusions in hematological
malignancies and recently in some types of epithelial cancer. Glioblastoma multiforme (GBM) is among the most lethal forms of human cancer. Here, an integrated gene fusion discovery pipeline was developed for the detection of in-frame fused transcripts from RNA- seq and genomic fusions from whole exome sequences. The application of the pipeline to human GBM unraveled recurrent chromosomal translocations, which fuse in-frame the tyrosine kinase domain of FGFR genes (FGFR l or FGFR3) to the TACC domain of TACC 1 or TACC3, respectively. The frequency of FGFR-TACC fusions is 3 of 97 GBM (3.1%). The FGFR-TACC fusion protein displays strong oncogenic activity when introduced, into astrocytes or transduced by lentivirus-medi ted stereotactic deiiveiy to the adult mouse brain. The FGFR-TACC fusion protein mis-localizes over the mitotic spindle pole, has constitutive tyrosine kinase activity and dysregulates the mitotic cycle with delayed mitotic progression. The impaired mitotic fidelity triggers chromatid cohesion defects, defective spindle checkpoint activation, chromosomal mis-segregation, and rampant aneuploidy. Inhibition of FGFR kinase corrects the aneuploidy and oral administration of a specific FGFR tyrosine kinase inhibitor under clinical investigation arrests tumor growth and prolongs survival of mice harboring intracranial FGFR3-TACC3-initiated glioma. FGFR-TACC fusions identify a subset of G BM patients who may benefit from targeted inhibition of the tyrosine kinase activity of FGFR.
[0(5253] Glioblastoma multiforme (GBM) is among the most difficult forms of cancer to treat in humans (Qhgaki and Kleihues, 2005). So far, the targeted therapeutic approaches that have been tested against potentially important oncogenic drivers in GBM have met limited success (Lo, 2010; Reardon et al., 2010; van den Bent et al., 2009). Recurrent chromosomal translocations leading to production of oncogenic fusion proteins are viewed as initiating and addicting events in the pathogenesis of human cancer, thus providing the most desirable molecular targets for cancer therapy (Ablain et al, 201 1 ; Mitelman et al., 2007).
Chromosomal rearrangements resulting in recurrent and oncogenic gene fusions are hallmarks of hematological malignancies and recently they have also been uncovered in subsets of solid tumors (breast, prostate, lung and colorectal carcinoma), but they have not been found in GBM (Bass et al., 201 1 ; Prensner and Chinnaiyan, 2009). Important and successful targeted therapeutic interventions for patients whose tumors carry these rearrangements have stemmed from the discovery of runctional gene fusions, especially when the translocations involve kinase-coding genes (BCR-ABL, EML4-ALK) (Druker, 2009; Gerber and Minna, 2010).
[ΘΘ254] A hallmark of GBM is rampant chromosomal instability (CIN), which leads to aneuploidy (Fumari et al, 2007). CIN and aneuploidy are early events in the pathogenesis of cancer (Cahill et al, 1999). It has been suggested that genetic alterations targeting mitotic fidelity might be responsible for mis-segregation of chromosomes during mitosis, resulting in aneuploidy (Gordon et al., 2012; Solomon et al., 201 1). Here, the first cases of recurrent and oncogenic gene fusions in human GBM are described. The resulting fusion protein localizes to mitotic cells, disrupts the normal control of chromosome segregation and induces aneuploidy. A therapeutic strategy with FGFR tyrosine kinase inhibitors is also reported for the targeted therapy of GBM patients harboring these chromosomal rearrangements.
[Θ0255] Identification of recurrent fusions of FGFR and TACC genes. To identify genomic rearrangements in GBM that generate functional fusion proteins and are recurrent, gene pairs discovered as in-frame fused transcripts from the analysis of massively parallel, paired-end sequencing of expressed transcripts (RNA-seq) would also emerge as fused gene pairs from the genomic analysis of hitman GBM. Towards this aim, two complementary gene fusion discovery methods were devised and were applied to two GBM cohorts. The first, TX-Fuse, is an algorithm for the discovery of candidate fusion transcripts from RNA- seq (Figure 8). The second, Exome-Fuse, detects fusion genes from whole exome DNA sequences (Figure 8). As first step for the detection ef fused transcripts, RNA-seq data was generated from short-term cultures of glioma stem-like cells (GSCs) freshly isolated fro nine patients carrying primary GBM. The culture of primary GBM tumors under serum-free conditions selects cells that retain phenotypes and genotypes closely mirroring primary tumor profiles as compared (o serum-cultured glioma ceil lines that have largely lost their developmental identities (Lee et al., 2006). Therefore, without being bound by theory, if glioma ceils cany gene fusions causally responsible for the most aggressive hallmarks of GBM, they should be selected in GSCs. RNA-seq generated an average of 60.3 million paired reads for each GSC culture, of which over 80% were mapped to (he reference transeriptome and genome. TX-Fuse detects two main sources of evidence: split reads and split inserts (see Experimental Procedures). The application of TX-Fuse to the RNA-seq dataset from nine GSCs led to the discovery of five candidate rearrangements (all of which were intrachromosomal) that give rise to in- frame fusion transcripts (Table 1).
[00256] Tab !e 1: Pre dieted in-ii -a me fusie si pre iteins from RNA -Seq of nine GSCs i serts f E-i iS Saw pis Seq 1 S c; 2 . .. Ί Pos2
GSC-
294 76 1 123 FGFR3 TACC3 NM_ _000142 _ 006342 2530 1751
GSC-
37 54 01 14 POLR2A WRAP53 NM_ _000937 _ 001 143990 479 798
GSC-
7 48 01 14 CAPZB UBR4 MM 001206540 N 020765 228 121 1 1
GSC-
8 29 0517 ST8SIA4 PAM MM 005668 NM 000919 125 730
GSC-
6 1 7 0308 P!GU COA6 NM 080476 NM 014071 729 6471
GSC-
1 6 IF AR2 !LI ORB NM 000874 000628 1083 149
#Sp>lit #Splii Chr Strand hg 19__. Chr Strand hg19 .
inserts Reacs Sample i 1 GenPosl 2 2 GenPos2
GSC-
294 76 1 123 4 + 1808842 4 + 1737004
GSC-
37 54 01 4 17 + 7399259 17 + 7604059
GSC-
7 48 01 4 1 - 19712098 1 - 19433440
GSC-
8 29 0517 5 - 100147809 5 + 102260661
GSC-
6 17 0308 20 - 33203914 20 - 33303130
GSC-
6 0127 21 + 34632901 21 + 34640699
[80257] Next, genomic rearrangements leading to gene fusions were identified in GBM by applying Exome-Fuse to a dataset of paired-end exome DN A sequences from 84 GBM samples from TCGA (Table 2). [0(5258] This analysis detected 147 paired gene fusions, thus producing an average of 1.75 gene fusion events per tumor (Table 3).
[60259] The FGFR and TACC families of genes were markedly enriched among those recurrently involved in genomic fusions, with eight tumors harboring FGFR rearrangements and seven tumors harboring fusions that implicate TACC genes (Figure 1A), The comparative analysis of the TX-Fuse and Exon-Fuse outputs revealed that FGFR3-TACC3 was the only fusion pair identified as either an in- frame transcript by TX-Fuse and genomic fusions by Exome-Fuse (Tables 1, 2 and 3).
[t>826(5] Table 2 shows fusion breakpoint information of recurrent gene fusions identified by Exome-fuse analysis of 84 GBM from TCGA. As multiple junctions may exist in each fusion candidate, information for all breakpoints is displayed. Column definitions include: sample - TCGA sample ID, virtForSplitReads/ virtRevSplitReads/virtTotSpiitReads - # forward/reverse/total split reads, spiitinserts = # split inserts, dirA/dirB = forward (1) or reverse (0) direction of split read portion mapping to gene A/B, dirABjtnatepair = direction of mate pair of spli t read, cosmicA+B = it recorded mutations of gene A+B in COSMIC.
Tabl ^.Fusion breakpoint information of recurrent gene fusions identified by Exome-fuse analysis of 84 GBM from TCGA.
TCGA-06-6390 10 9 19 8 FGFR3 c r4 + 1778521 TACC3 chr4 + 1708787 1 0 2803
TCGA-12-0826 5 6 11 5 FGFR3 chr4 + 177S502 TACC3 chr4 + 1707185 0 0 1 2803
TCGA-19-595S 3 0 3 2 FGFR3 chr4 + 1778539 TACC3 chr4 + 1707203 0 1 1 2803
TCGA-27-1835 11 1 12 4 FGFR3 chr4 + 1778595 TACC3 chr4 + 1709397 0 0 1 2803
TCGA-12-0820 7 2 9 4 FGFR3 chr4 + 1779184 PRKG2 chr4 - 82338347 1 1 0 2805
TCGA-12-1038 3 1 4 4 ABL1 chr9 + 132597569 TNFRSFIOB chrS - 22936252 0 0 1 892
TCGA-06-1802 7 1 8 8 ADA 12 chrlO - 127698245 PTPRD chrS - 8596127 0 0 1 54
TCGA-06-1801 7 0 7 5 HIP1 chr7 - 75010010 PTPRD chr9 - 9387093 1 0 0 52
TCGA-12-1088 3 0 3 3 KIDINS220 chr2 - 88S6300 PPP1R3A chr7 - 113305567 0 0 1 45
TCGA-12-1088 37 1 38 10 IDINS220 chr2 - 8887075 PPP1R3A chr7 - 113305191 1 1 0 45
TCGA-32-2491 2 17 19 6 ODZ1 chrX - 123342503 STAG 2 chrX + 123019118 0 1 0 36
TCGA-32-2491 11 1 12 10 ODZ1 chrX - 123526882 5ASH3 chrX + 128749198 1 0 0 34
TCGA-12-0829 24 0 24 13 LRRK2 chr!2 + 39032542 V5NL1 chr2 + 17630556 1 1 0 32
TCGA-12-0829 25 1 25 13 L RK2 chrl2 + 38975444 VS L1 chr2 + 17639377 1 0 0 32
TCGA-12-0829 87 16 103 58 LRR 2 chrl2 + 38975652 V5NL1 chr2 + 17639552 0 1 1 32
TCGA-13-0957 3 2 5 6 NUDT19 chrl9 + 37891921 ODZ1 chrX - 123925223 0 0 32
TCGA-12-1088 12 i 13 5 GLI3 chr7 - 420313S0 RIMBP2 chrl2 - 129517282 1 0 0 31
TCGA-12-1088 5 0 5 1 GLI3 chr7 - 42031574 RI BP2 chrl2 - 129517455 0 1 1 31
TCGA-12-1089 10 0 10 5 AHNAK chrll - 62056459 C21orf29 chr21 - 44923276 0 1 1 30
TCGA-06-1801 27 1 28 12 CROCC chrl + 17171362 CSMD2 chrl - 34381139 1 0 0 29
TCGA-12-1089 12 1 13 6 CLK3 chrlS + 72705401 LRP1 chrl2 + 55S80002 0 1 1 28
TCGA-12-1089 14 2 16 8 CL 3 chrlb + 72705248 LRP1 chrl2 + 55879646 1 0 0 28
TCGA-12-1089 42 5 47 24 LA A2 c r6 + 129836071 PDE10A chr6 - 165858426 1 0 0 28
TCGA-05-1802 48 9 57 27 LAMA2 chr6 + 129483265 5EC14L3 chr22 - 29193005 1 1 0 27
TCGA-19-0957 4 16 20 6 CSMD2 chrl - 34115076 DH2 chr7 + 75525221 0 0 1 27
TCGA-06-1801 21 1 22 4 FA 192A chrl6 - 55757701 LRP1 chrl2 + 55858598 0 0 1 26
TCGA-12-1089 27 0 27 2 FGFR4 chrS + 176447670 L!LRBl chrl9 + 59840807 Λ 0 0 25
TCGA-19-0957 0 I 4 EML1 chrl4 + 99349006 NRXN3 chrl4 + 79233969 1 0 0 24
TCGA-06-1801 19 133 152 51 NHS!.2 chrX + 71082676 TAF1 chrX + 70520522 0 0 22
TCGA-06-1801 51 3 54 8 NHSL2 chrX + 71083319 TAF1 chrX + 70521607 1 0 1 22
TCGA-12-1089 9 0 9 4 CACNA1C chrl2 + 2325330 ITGAV chr2 + 187195411 0 0 1 22
TCGA-19-0957 8 1 9 6 CDH11 chrlS - 63579650 RERE chrl - 8588774 0 0 1 22
TCGA-12-0829 12 3 15 4 ENTPD2 chrS - 139062591 FREM2 chrl3 + 38318644 1 1 0 21
TCGA-12-0829 3 5 1 EFS chrl4 - 22896776 XM3 chrl4 + 78678529 1 0 1 21
Tabif- 7< continuation
TCGA-12-0829 56 6 62 14 DiS3L chrlS 64377566 GLI3 chr7 - 42032535
TCGA-12-0829 8 2 10 3 EFS chrl4 22896431 NRXM3 chi-14 ÷ 78678139
TCGA-12-0829 9 65 74 37 DfS3L chrlS 64377398 GU3 chr7 - 42032341
TCGA-27-1S35 14 0 14 4 FA 19A2 chr!2 60707200 GU I chrl2 ÷ 56146523
TCGA-06-1801 20 0 20 2 FREM2 chrl3 38163882 RALYL chrS ÷ 85785432
TCGA-12-0827 2 0 2 2 A8CC12 chr!6 46722685 FGFR4 chrS + 176457194
TCGA-12-0829 35 0 35 7 ANXA7 chrlO 74808655 CACNAIC chrl2 ÷ 2458351
TCGA-06-2559 60 37 97 1 PLE HM3 chr2 208426920 PTPRS chrl9 - 5222592
TCGA-12-1088 2 0 2 2 PLCL1 chr2 198630224 TACC2 chrlO + 123987513
TCGA-06-1801 10 0 10 4 FGFR4 chr5 176450528 WiSP2 chr20 ÷ 42782576
TCGA-06-1802 15 0 15 2 PDHA2 chr4 96980717 PDZRN4 chrl2 + 39959553
TCGA-06-1802 4 0 4 2 PDHA2 chr4 96980509 PDZRN4 chrl2 ÷ 39959384
TCGA-06-6390 53 0 53 18 GPR182 chrl2 55675639 PDZRN4 chrl2 ÷ 39957003
TCGA-12-0829 1121 252 1373 602 ADCY8 chr8 131886108 SSX3 chrX - 48091929
TCGA-12-0829 14 8 22 3 ADCY8 chr8 131886506 SSX3 chrX - 48091719
TCGA-12-0829 9 42 51 18 ADAM 12 chrlO 127733231 DAPKl chr9 ÷ 89454764
TCGA-12-3653 22 0 22 10 JOSD2 chr!9 55705579 PTPRS chrlS - 5245999
TCGA-12-0829 100 0 100 20 COL14A1 chr8 121370990 MP12 chrll - 102242881
TCGA-12-0829 152 0 152 24 COL14A1 chrS 121371195 MMP12 chrll - 102242953
TCGA-06-1802 11 47 58 19 MUSK chr9 112509906 SYNP02 chr4 + 120172123
TCGA-06-1805 6 4 10 6 COL14A1 chr8 1213320S0 NCRNA0015 chi-21 - 18174873
TCGA-12-0822 37 0 37 3 C7orf44 chr7 43683128 TACC2 chrlO + 123835337
TCGA-12-0829 0 2 2 365 GSTA3 chr6 52878492 TACC2 chrlO ÷ 123884543
TCGA-12-0829 124 16 140 51 GSTA3 chr6 52878680 TACC2 chrlO ÷ 123884705
TCGA-12-0829 21 7 28 10 HfPl chr7 75022909 MASPl chr3 - 1884S2372
TCGA-12-0829 268 123 391 242 H!Pl chr7 75022741 MASPl chr3 - 188452581
TCGA-12-0829 36 641 677 365 6STA3 chr6 52878496 TACC2 chrlO + 123884531
TCGA-12-1088 10 1 11 3 CAMTA1 chrl 7710762 TMPRSS3 chr21 - 42665918
TCGA-12-1088 65 0 65 6 ADCY10 chrl 166139873 DUSP27 chrl ÷ 165351555
TCGA-12-1088 8 1 9 4 CA TA1 chrl 7714539 TMPRSS3 chr21 - 42666044
TCGA-27-1S35 83 1 84 22 CMYA5 chrS 79120729 SRRM1 chrl ÷ 24870899
TCGA-06-1801 0 43 43 31 CAMTA1 chrl 7264935 GDPD2 chrX ÷ 59563759
TCGA-06-1801 13 41 54 31 CAMTA1 chrl 7265429 GDPD2 chrX + 69563431
TCGA-06-1801 24 66 90 61 CAMTA1 chrl 7265556 GDPD2 chrX ÷ 69563762
- y
T«b!e 7 continuation
TCGA-12-0829 2 0 3 CCDC147 chflO + 106165013 iSX chr22 33795708 0 1
TCGA-12-1088 7 1 8 5 C YA5 chr5 + 79045621 STK24 chrl3 97969547 1 0
TCGA-06-1801 1 8 4 DEPDC5 chr22 + 30619774 ROBOi chr3 - 79802538 0 1
TCGA-12-0820 110 20 130 23 ABCA13 chr7 + 48597322 NHSL2 chrX ÷ 71077547 1 0
TCGA-12-0820 29 4 33 3 ABCA13 chr7 + 48597477 HSL2 chrX 71077690 0 1
TCGA-12-0829 46 2 48 4 Ui\!9 chrl - 224536835 COR1 chr!7 15883585 0 0
TCGA-12-3644 3 0 3 1 EFHC1 chr6 + 52432073 LRBA chr4 - 151418615 1 0
TCGA-12-3644 3 10 13 3 EFHC1 chr6 + 52431890 LRBA chr4 - 151418438 1 0
TCGA-19-5958 6 6 12 7 DEPDC5 chr22 + 30504095 SLC5A4 chr22 - 30974671 0 1
TCGA-06-1801 4 4 8 5 KCND3 chrl - 112227957 LY75 chr2 160443238 1 1
TCGA-12-0820 26 1 27 2 BBX chr3 + 108997451 CUL3 chr2 - 225108623 0 0
TCGA-12-0828 8 67 75 31 ADCY2 chr5 + 7558840 SDAD1 chr4 - 77096208 1 1
TCGA-12-0829 13 21 34 16 AGBL4 chrl - 48902776 NUP188 chr9 + 130808425 0 0
TCGA-12-0829 64 308 372 197 EY5 chr6 - 64513356 iLIRN chr2 + 113603712 0 1
TCGA-12-0829 7 25 32 11 A.GBL4 chrl - 48902600 NUP18S chr9 ÷ 130808628 1 1
TCGA-12-0829 9 0 9 1 LR3A chr4 - 151790893 PSENl chr!4 + 72707509 1 0
TCGA-12-1093 65 4 69 21 OSBPL10 chr3 - 31687272 TRAPPC9 chr8 - 140828099 1 1
TCGA-12-1600 9 0 9 5 5-Sep chr22 + 18088018 NC0R1 chrl7 15915170 0 1
TCGA-19-0957 18 1 19 7 ADCY10 chrl - 166060645 A T3 chrl - 241743142 0 1
TCGA-19-0957 34 2 36 11 ADCY10 chrl - 166060502 AKT3 chrl - 241742588 1 0
TCGA-12-0822 0 1 1 16 ITGB2 chr21 - 45147805 SH3RF3 chr2 + 109430489 0 1
TCGA-12-0822 6 2 8 1 ITGB2 chr21 - 45147994 SH3RF3 chr2 + 109430669 0 1
TCGA-12-0827 25 3 28 8 CUL3 chr2 - 225126210 LY75 chr2 - 160455052 1 0
TCGA-12-0828 7 2 9 4 FH chrl - 239743589 SRGAPl chr!2 + 62723692 0 1
TCGA-12-0829 24 0 24 9 ITGA9 chr3 + 37712050 SNX5 chr20 - 17885523 0 1
TCGA-12-1089 17 2 19 5 ABCC1 chrl6 + 16077635 RNF216 chr7 5692038 1 1
TCGA-12-1089 6 0 6 8 CAMSAP1 chr9 - 137867066 NCF2 chrl 181799323 1 0
TCGA-19-0957 16 0 16 4 CCDC147 chrlO + 106114657 STK4 chr20 43111359 0 0
TCGA-06-1801 5 33 38 18 AP4S1 chrl4 + 30611930 EYS chr6 - 64770011 1 0
TCGA-06-1S05 3 14 17 9 CUL3 chr2 - 225064315 SLC44A2 chr!9 + 10608393 1 0
TCGA-12-0829 14 27 41 23 ADCY2 chrS + 7798046 C14orfl74 chrl4 ÷ 76914809 1 0
TCGA-12-0829 59 7 66 18 NR3C1 chr5 - 142760085 SORCS2 chr4 + 7354165 0 0
TCGA-12-0829 9 40 49 28 ADCY2 chr5 + 7798641 C14orfl74 chr!4 76915034 0 1
TCGA-12-1093 20 0 20 4 GAPVD1 chr9 + 127104266 MAPKAPl chr9 127490362 1 0
Tishle 7· continuation
TCGA-12-1600 7 0 7 4 C!LP chrl5 - 63283865 PARP16 chrl5 63350048
TCGA-19-0957 13 3 15 6 AQP2 chrl2 + 48635567 CDH4 chr20 + 59413648
TCGA- 19-0957 6 0 6 1 AQP2 chrl2 + 48635406 CDH4 chr20 + 59413468
TCGA-06-0166 2 0 2 3 CCDC158 chr4 - 77541796 SNX5 chr20 17885346
TCGA-06-1802 30 0 30 9 RANBP2 chr2 + 108758804 SATB2 chr2 199895572
TCGA-06-1805 4 0 4 3 C2CD3 chrli - 73430819 XRRA1 chrli - 74309669
TCGA-06-1805 6 1 7 5 NEUROG1 chr5 - 134898853 PRKCH chrl4 + 61027580
TCGA-12-0820 27 0 27 2 RANBP2 cht-2 + 103749908 TTC27 chr2 + 32839367
TCGA- 12-0820 58 7 65 15 RANBP2 chr2 + 108749412 TTC27 chr2 + 32837790
TCGA-12-0829 6 128 134 35 C2CD3 chrli - 73529639 CAPZB chrl 19556435
TCGA- 12-0829 84 443 527 227 C2CD3 chrli - 73529293 CAPZB chrl - 19556627
TCGA-12-10S8 10 0 10 2 P ACS INI chr6 + 34589431 TNC chr9 - 116884742
TCGA-12-1088 12 0 12 2 PACSI 1 chr6 + 34589619 TNC chr9 - 116884958
TCGA-19-0957 34 19 53 17 PRKCH chrl4 + 61032978 ZFAND3 chr6 + 3822S111
TCGA-19-0957 7 1 8 7 APKAP1 chr9 - 127348507 SLC9A1 chrl - 27302334
TCGA-19-0957 8 39 47 21 PRKCH chrl4 + 61032774 ZFAND3 chr6 + 38227949
TCGA-06-1801 5 11 16 4 MAOA chrX + 43486192 SH3RF3 chr2 + 109237058
TCGA-06-1802 10 12 22 12 DNM1L chrlz + 32736794 SYNP02 chr4 + 120172271
TCGA-06-1802 18 42 60 24 MUC4 chr3 - 196982875 SM0C2 chr6 + 168676813
TCGA-12-0329 6 0 6 6 AT 1 chr6 - 16669201 CACNAIG chrl7 + 46004995
TCGA-12-0829 7 0 7 3 ATP6VOD2 chr8 + 87186716 RERE chrl - 8336574
TCGA-12-1088 11 1 12 6 BCAS3 chrl7 + 56321892 CACNAIG chrl7 + 46010698
TCGA-12-1088 15 2 17 5 ABCC1 chrl6 + 16135771 AGBL4 chrl - 49315120
TCGA-12-1088 17 3 20 4 MST1R chr3 - 49910627 WDFY1 chr2 - 224512774
TCGA-12-1088 4 0 4 2 FBXL4 chr6 - 99431443 SYNP02 chr4 + 120172560
TCGA-12-1092 39 4 43 12 CNTN2 chrl + 203302926 DNAJC6 chrl + 65591195
TCGA-12-1598 4 0 4 5 MPP1 chrX - 153673715 SRGAPl chrl2 + 62777947
TCGA-19-17S6 5 19 24 7 ATP5B chrl2 - 55320148 USP48 chrl 21920103
TCGA-19-2621 21 0 21 3 BCAS3 chrl7 + 56731673 TTYH1 chrl9 + 59638801
TCGA-06-1801 15 0 15 9 ClSorf23 chrl5 + 38469150 DMD chrX - 32092185
TCGA-06-1805 6 3 9 5 FAM19A2 chrl2 - 60547321 POLM chr7 44082653
TCGA-12-0829 13 84 97 44 ATP5B chrl2 - 55318484 PRC1 chrl5 - 89330475
TCGA-12-0829 158 207 365 44 ATP SB chrl2 - 55320850 PRC1 chrl5 89334458
TCGA-12-0829 2 1 3 2 DDI2 chrl + 15825507 IDINS220 chr2 8805399
Tishle 7, continuation
TCGA- -12-0829 25 6 31 44 ATP5B chrl2 - 55321832 PRC1 chrl5 - 89335627 1 0 1 7
TCGA- •12-0829 34 21 55 44 ATP5B chrl2 - 55321200 PRC1 chrl5 - 89335044 0 1 0 7
TCGA- 12-0829 44 6 50 4 A3CC6 chrl6 - 16204784 SUMF1 chr3 - 4470138 1 0 0 7
TCGA- -12-0829 53 28 81 35 DDI2 chrl + 15825941 KIDIN5220 chr2 - 8805580 1 0 1 7
TCGA- 12-0829 9 0 9 5 D D chrX - 32013100 N4BP2L2 chrl3 - 32008512 0 0 1 7
TCGA- -12-1092 a 0 9 4 LRRC4B chrl9 - 55754780 NR3C1 chr5 - 142660156 0 1 0 7
TCGA- 19-2621 3 22 25 11 PCDH12 chr5 - 141309153 SLC36A2 chr5 - 150679274 1 1 0 7
TCGA- ■06-1802 8 4 12 6 3AHD1 chrlS + 38539023 OSBPLIO chr3 - 31729622 0 1 1 6
TCGA- 12-0828 11 0 11 1 PLOD3 chr7 - 100646340 VSNLl chr2 + 17638618 1 1 0 6
TCGA- 12-0828 40 9 49 20 PLODS chr7 - 100646511 VSNLl chr2 + 17637955 0 0 1 6
TCGA- -12-0829 16 2 18 g C21orf29 chr21 - 44922864 MYT1 chr20 + 62300829 0 0 1 6
TCGA- -12-0829 196 0 196 37 IGFBP3 chr7 - 45922866 SMOC2 chr6 + 168722450 0 0 1 6
TCGA- -12-0829 5 1 6 1 FAM168A chrll - 72839771 NCF2 chrl - 181826115 1 0 1 6
TCGA- -12-0829 5 18 23 9 FAM168A chrll - 72839534 NCF2 chrl - 181825930 1 0 0 6
TCGA- -12-1089 20 0 20 2 SLC44A2 chrl9 + 10602997 XRCC4 chr5 + 82430803 1 0 0 6
TCGA- ■19-0957 17 1 18 4 PAX3 chr2 - 222778052 WDFY1 chr2 - 224453159 1 0 1 6
TCGA- -06-1801 5 0 5 1 CAP2 chr6 + 17571234 DNAJC6 chrl + 65602700 1 0 0 5
TCGA- 06-1801 6 32 38 15 CAP2 chr6 + 17571666 DNAJC6 chrl + 65603089 0 1 1 5
TCGA- 06-1805 3 0 3 8 PLCLl chr2 + 198578552 SURF6 chr9 - 135188818 0 1 0 5
TCGA- 06-1805 7 2 9 4 PLCLl chr2 + 198578671 SURF6 chr9 - 135189294 1 0 1 5
TCGA- 12-0822 17 4 21 4 TAAR6 chr6 + 132933266 TTYH1 chrl9 + 59629451 0 1 1 5
TCGA- 12-0828 17 0 17 8 AQP2 chrl2 + 48634800 ECE1 chrl - 21515240 0 1 1 5
TCGA- 12-0828 7 0 7 1 AQ.P2 chrl2 + 48634610 ECE1 chrl - 21515033 1 0 0 5
TCGA- 12-0829 12 0 12 7 CACNA1G chrl7 + 46039372 CNTNAP4 chri6 + 74873868 0 1 1 5
TCGA- ■19-0957 4 0 4 3 PCDH12 chrS - 141316624 SH3BP5 chr3 - 15315567 0 0 1 5
TCGA- 19-0957 8 2 10 2 PCDH12 chrS - 141316405 SH3BP5 chr3 - 15315731 1 1 0 5
TCGA- 06-1801 13 1 14 1 ABCC6 chrl6 - 16205051 CMT 7 chr3 + 32443880 0 1 1 4
TCGA- ■06-1801 33 7 40 4 ABCC6 chrl6 - 16204860 C T 7 chr3 + 32443722 1 0 0 4
TCGA- 06-1805 10 3 13 6 AGBL4 chrl - 49449813 NOX4 chrll - 88714996 0 0 1 4
TCGA- 12-0829 11 0 11 4 FA 160A1 chr4 + 152595916 LY75 chr2 - 160440194 0 1 0 4
TCGA- ■12-0829 17 1 18 5 FAM160A1 chr4 + 152596097 LY75 chr2 - 160440376 1 0 1 4
TCGA- -12-0829 487 83 570 249 COR07 chrl6 - 4375428 DYR 3 chrl + 204876154 0 0 1 4
TCGA- •12-10SS 2 12 14 4 FA 172A chrS - 93052315 TRIOBP chr22 + 36427382 0 1 1 4
TCGA- -06-1801 30 7 37 18 DEPDC7 chrll + 33003811 EIF2C2 chrS - 141618836 1 0 0 3
T'nbie / continuation
TC6A-06-1801 40 28 68 33 MAP7 chr6 - 136728609 SH3RF3 chr2 + 109392677 0 0
TCGA-12-1093 6 15 21 8 COR07 chrl6 - 4398302 PLEK2 chrl4 66934201 0 1
TCGA-12-3644 33 0 33 4 EDA chrX + 69073054 5SX3 chrX - 48094443 1 1
TCGA-12-3644 37 15 52 17 C15orf33 chrl5 - 47424122 PARP16 chrlS - 63350289 0 0
TCGA-19-1791 14 3 17 8 PSEN1 chrl4 + 72748293 ZNF410 chrl4 + 73431112 0 0
TCGA-06-1802 35 2 37 17 CELF2 chrlO + 11352537 PLA2G2F chrl + 20348173 1 1
TCGA-06-1802 63 26 89 25 CELF2 chrlO + 11352765 PLA2G2F chrl + 20347997 0 0
TCGA-06-1802 S 2 10 8 LCLAT1 chr2 + 30535977 PACSINl chi-6 + 34576195 1 0
TCGA-06-2562 6 0 6 4 SNTA1 chr20 - 31473415 TME 80 chrll + 689744 0 0
TCGA-12-0829 16 1 17 4 LASS6 chr2 + 169045211 (MKAI 2 chr6 + 125021252 0 0
TC6A-12-0829 7 0 7 2 LASS6 chr2 + 159045333 NKAIN2 chrb + 125021072 1 1
TCGA-14-0813 339 39 378 5 SNTA1 chr20 - 31481069 TMEM80 chrll + 686739 0 0
TC6A-12-0820 49 3 52 11 CAM K1 chrl7 - 3712344 FA 184B chi-4 - 17271273 0 1
TCGA-12-0826 8 18 26 13 CELF2 chrlO + 11406463 N E4 chrl6 + 389427 1 1
TCGA-12-1089 17 4 21 10 C6orfl70 chr6 - 121478035 NKAIN2 chr6 + 125083380 1 0
TCGA-12-1600 19 0 19 3 ATP6AP1L chr5 + 81649744 FAM172A chr5 - 93336459 1 0
TCGA-12-1600 5 35 40 6 ATP6AP1L chr5 + 81649902 FA 172A chr5 93336676 1 0
TCGA-19-1790 4 0 4 4 ARMC6 chrl9 + 19026932 FAM184B chi'4 - 17391210 1 0
TCGA-06-1802 12 0 12 8 EIF2C2 chr8 - 141648334 TNFRSFIOB chr8 - 22940680 0 0
TCGA-14-0781 22 2 24 8 FAM160A1 chr4 + 152584637 UNC93B1 chrll 67523253 1 0
I'able 3 Recurrent gene fusion pairs from Exome-fuse analysis of 84 GBM from TCGA.
Sample gene A gene B Sample gene A gene B
TCGA-12-0820 ABCA13 NHSL2 TCGA-12-0820 CAMKKl FA 184B
TCGA-12-10S9 ABCC1 RNF216 TCGA-12-1089 CAMSAPl CF2
TCGA-12-10S8 ABCC1 AGBL4 TCGA-12-1088 CAMTAl TMPRSS3
TCGA-12-0827 ABCC12 FGF 4 TCGA-06-1801 CAMTAl GDPD2
TCGA-12-0829 A8CC5 SU F1 TCGA-06-1801 CAP 2 DNAJC6
TCGA-06-1801 A8CC6 C TM7 TCGA-19-0957 CCDC147 ST 4
TCGA-12-108S A8L1 TNFRSF10B TCGA-12-0829 CCDC147 !SX
TCGA-06-1802 ADASV112 PTPRD TCGA-06-0166 CCDC158 5NX5
TCGA-12-0829 AD AMI 2 DAPK1 TCGA-19-0957 CDH11 RERE
TCGA-12-1088 ADCY10 OUSP27 TCGA-06-1802 CELF2 PLA2G2F
TCGA-19-0957 ADCY10 A T3 TCGA-12-0826 CELF2 NME4
TCGA-12-082S ADCY2 SDAD1 TCGA-12-1600 CSLP PARP16
TCGA-12-0829 A0CY2 C14orfl74 TCGA-12-1089 CLK3 LR.P1
TCGA-12-0829 ADCY8 SSX3 TCGA-12-1088 C YA5 STK24
TCGA-12-0829 AG8L4 NUP188 TCGA-27-1835 CMYA5 SRRM1
TCGA-06-1805 AGSL4 OX4 TCGA-12-1092 CNT 2 DNAJC6
TCGA-12-1089 AH AK C21orf29 TCGA-06-1805 COL14A1 CRNA00157
TCGA-12-0829 A X.A7 CACNA1C TCGA-12-0829 COL14A1 MP12
TCGA-06-1801 AP4S1 EYS TCGA-12-1093 COR07 PLEK2
TCGA-12-0828 AG.P2 ECE1 TCGA-12-0829 COR07 DYRK3
TCGA-19-0957 AG.P2 CDH4 TCGA-06-1801 CROCC C5 D2
TCGA-19-1790 ARMC6 FAM184B TCGA-19-0957 CSMD2 MDH2
TCGA-19-1786 ATP5B USP48 TCGA-06-1805 CUL3 SLC44A2
TCGA-12-0829 ATP5B PRC1 TCGA-12-0827 CUL3 LY75
TCGA-12-1600 ATP6AP1L FA 172A TCGA-12-0829 DDi2 KIDINS220
TCGA-12-0829 ATP6V0D2 RERE TCGA-19-5958 DEPDC5 SLC5A.4
TCGA-12-0829 ATX l CACNAIG TCGA-06-1801 DEPDC5 ROBOl
TCGA-06-1802 BAHD1 OSBPL10 TCGA-06-1801 DEPDC7 EiF2C2
TCGA-12-0820 BBX CUL3 TCGA-12-0829 OiS3L GU3
TCGA-19-2621 BCAS3 ΤΤΎΗ1 TCGA-12-0829 OMD N4BP2L2
TCGA-12-1088 BCAS3 CACNAIG TCGA-06-1802 DNM1L 5YNP02
TCGA-06-1801 C15orf23 DMD TCGA-12-3644 EDA SSX3
TCGA-12-3644 C15orf33 PA P16 TCGA-12-3644 EFHC1 LRBA
TCGA-12-0829 C21orf29 MYT1 TCGA-12-0829 EFS RX 3
TCGA-06-1805 C2CD3 XRRA1 TCGA-06-1802 EIF2C2 T FRSF10B
TCGA-12-0829 C2CD3 CAPZB TCGA-19-0957 EML1 NRX 3
TCGA-12-1089 C6orfl70 KAIN2 TCGA-12-0829 ENTPD2 FREM2
TCGA-12-0822 C7orf 4 TACC2 TCGA-12-0829 EYS 1L1R
TCGA-12-1089 CACNA1C iTGAV TCGA-14-078 FAM160A1 UNC93B1
TCGA-12-0829 CACNAIG C T AP4 TCGA-12-0829 FAM150.A1 LY75
[80201 ] Table 3 above shows recurrent gene fusion pairs from Exome-fuse analysis of 84 GBM from TCGA. Fusion candidates have been nominated if they have at least two split inserts and at least two split reads. To further filter the list on recurrence, any fusion candidate was kept in which one of the genes is involved in at least two fusions across different samples.
[00262] To experimentally validate the computational predictions that emerged from TX- Fuse, the PGR products spanning the fusion breakpoint were sequenced and validated each of the five in-frame fusion predictions (Figures 1 and 9). In Figure IB, the prediction is shown and in Figure 1C, the cDNA sequence validation for the fusion with the highest read support involving FGFR3 fused in- frame with TACC3 in GSC-1 123 is shown. The same FGFR3- TACC3 fusion transcript was also detected in the primary GBM- 1123 tumor specimen from which the GSC-1 123 culture was established (Figure 1C). The amplified cDMA contained an open reading frame for a protein of 1 ,048 amino acids resulting from the fusion of a FGFR3 arnino-terminal portion of residues 1-758 with a TACC3 carboxy-terminal portion of residues 549-838 (Figure ID). FGFR3 is a member of the FGFR receptor tyrosine kinase (TK) family that transduces intracellular signals after binding to FGF iigands (Turner and Grose, 2010). TACC3 belongs to the evoiutionarily conserved TACC gene family, which also includes TACC l and TACC2. The distinctive feature of TACC proteins is the presence of a coiled-coil domain at the C-terminus, known as the TACC domain. Through the TACC domain, 'TACC proteins localize to the mitotic spindle during metaphase and stabilize the microtubule spindle network (Hood and Royle, 201 1; Peset and Vernos, 2008). In the predicted fusion protein the intracellular TK domain of FGFR3 is fused upstream of the TACC domain of TACC3 (Figure ID).
[80263] Exon-specific gene expression analysis from the RNA-seq coverage in GSC-1 12.3 demonstrated that the FGFR3 and TACC3 exons implicated in the fusion are highly overexpressed compared with the rnRNA seq ences not included in the fusion event (Figure IDA). Quantitative RT-PCR showed that the expression of the fused FGFR3-TACC3 exons is significantly higher in GSC-1 123 than other GSCs and the normal brain (80 to 130- fold, Figure 10B). Without being bound by theory, functionally significant genetic rearrangements may result in marked overexpression (outlier) of the genes implicated in the fusion events (Tomiins et al, 2007; Tomlins et al, 2005). The FGFR3-TACC3 fission protein was also abundantly expressed in GSC-1 123 and in the primary tumor GBM-1 123, as shown by Western blot and immunohistochemistTy (Figures IOC and 10D). On a Western Blot, the FGFR3-TACC3 fusion protein migrated at a size of -Ί50 kD and immunoprecipitation followed by mass spectrometry revealed the presence of FGFR3 and TACC 3 peptides consistent with the cDNA translation prediction (Figure IDE). Using PGR, the genomic breakpoint coordinates were mapped to chromosome 4 (#1,808,966 for FGFR3 and
#1,737,080 for TACC3, genome build GRCh37/hgl9) failing within FGFR3 exon 17 and TACC3 intron 7, which gives rise to a transcript in which the 5' FGFR3 exon 16 is spliced to the 3 ' TACC3 exon 8. The DN A junctions of FGFR3 and TACC3 show microhomology within a 10-base region, an observation consistent with results previously reported for other chromosomal rearrangements in hitman cancer (Bass et aL, 2.01 1 ; Stephens et al, 2009) (Figure IE).
[80264] The experimental validation of the inferred genomic fusions was focused on FGFR3-TACC3. Exome-Fuse identified FGFR3-TACC3 gene fusions in four GBM samples with breakpoints spanning invariably within intron 16 of FGFR3 (which is downstream to the coding region for the TK domain) and intron 7- 10 of TACC3 (which is upstream to the TACC domain) (Figure 2 A, Tables 4 and 5 ). Among the four positive TCGA GBM specimens, two were available from TCGA centers for molecular analysis (TCGA-27-1835 and TCGA-06-6390) and, by Sanger sequencing, each of them were confirmed to carry an in- frame fusion transcript that is consistent with the predicted genomic breakpoints (Figures 2B and 2C). Thus, the frames of the FGFR3-TACC-3 fusion proteins invariably result in juxtaposing the TK domain of FGFR3 upstream of the TACC domain of TACC3. Consistent with the abundant expression of FGFR3-TACC3 in GSC-1123 and GBM- 1123, the niRNA expression analysis of the TCGA tumors revealed that the four FGFR3-TACC3-posirive GBM display marked co-outlier expression of FGFR3 and TACC3 (Figure 2D). Recurrent gene fusions can be associated with local copy number variations (CNV) of the breakpoint regions (Wang et al., 2009). Accordingly, the analysis of SNP arrays in the TCGA dataset revealed the presence of microampiification events of the FGFR3 and TACC3 genes in all four FGFR3-TAC-C3-positive GBM (Figure 2E).
Table 4: List of split inserts supporting the identification of FGFR3-TACC3 fusion genes in four GBM samples from the ATLAS-TCGA exome collection
trrmr
aces
3 «<
-f ti
5 «
3 is s ¾ & ¾ ;a s
3 8
- 101 - Table 5 (Cont)
{00265] The FGFR3 and TACC3 genes are located 48-Kb apart on human chromosome 4 l 6. The other members of the FGFR and TACC families retain the close physical association of FGFR3 and TACC3, with FGFR1 and TACC1 paired on chromosome 8pl l and FGFR2 and TACC2 paired on chromosome 1 Oq'26. Without being bound by theory, the ancestral FGFR and TACC genes were physically linked and that this tandem gene cluster was duplicated at least twice to generate the FGFR1-TACC1, FGFR2-TACC2 and FGFR3- TACC3 pairs thai mark mammalian evolution (Still el al., 1999), The highly conserved TK domains among FGFR genes and TACC domains among TACC genes together with their invariable fusion in the FGFR3-TACC3 rearrangements prompted to ask whether other intra- chromosomal FGFR-TACC fusion combinations exist in human GBM.
[ΘΘ266] cDNA from a panel of 88 primary GBM were screened using pairs of upstream PGR primers thai bind the amino-terminal coding region of the TK domains of FGFRl, FGFR2 and FGFR3 and downstream primers that bind to the carboxy-tenninal coding region of the TACC domains of TACC 1 , TACC2 and TACC3 genes, respectively. The screening resulted in the identification of intrachromosomal FGFR-TACC fusions in two additional cases (one harboring FGFRl -TACC 1 and one FGFR3-TACC3), corresponding to three of 97 total GBM (3.1%), including the GBM- 1 123 case. The FGFRl -TACC 1 fusion breakpoint in GBM-51 joined in- frame exon 17 of FGFR l to exon 7 of TACC 1, resulting in a novel protein in which the TK domain of FGFRl is fused upstream of the TACC domain of TACC 1 (Figure 2F). The same structure was conserved again in GBM-22 in which exon 6 of FGFR3 is joined in-frame to exon 10 of TACC3 (Figure 2G). None of the tumors harboring FGFR-TACC fusions had mutations in IDHl or IDH2 genes, thus indicating that FGFR- TACC-positive GBM mark an independent subgroup of patients from those carrying IDH mutations (Table 6) (Yan et al., 2009). The constant linkage of the FGFR-TK to the 'TA CC domain created in each of the seven GBM harboring FGFR-TACC rearrangements suggests thai FGFR-TACC fusion proteins may generate important functional consequences for oncogenesis in the brain.
[ 0267] Table 6.
Age at initial (DH1-2 status iDHI-2 statas
Samples T¾?pe Time Status
athoiogic diagnosis (Sanger) (sxome)
TCGA-12-0826 FGFR3-TACC3 345 DECEASED 33 WT WT
TG6ArZ?-1835 FGFR3-TACC3 DECEASED HA WT
TCGArlS-SSSB FGFR3-TAGC3 164 LSVSNG 56 HA WT
TCGA Oe-6390 FGFR3-TAGC3 163 DECEASED 58 WT WT
GBM-22 FGFR3-TACC3 393 DECEASED WT m
GB - 123 FGFR3-TACC3 DECEASED W m
GBM-51 FGFR 5 -TACC': m HA m W:T m
Time = Survival ( days after diagnosis)
Sarsge? = ai¾alys¾s fi !ie by Sanger sequencing of geriomic DNA
Exon* = a&iaS sis done by 8» SAVI (Siafedcal AlgoraSro fc? ariant fceniificasion}, an a¾os»im
dev-atoped to ttetect point m& m cancer (BRAF Mutaiions Hary-Cei; Lsufemia. liacd E et at
The fifewEngand J¾t;msi: of Medicine 2011 A 16;36 (24):23e5-15)
fi¾= ^ Available
WT = tkS < T» sequence for R132 and 172 of sDH i and fDH2, respecfee!y [0(5268] Transforming activity of FGFR-TACC fusions. To test the functional importance of the FGFR-TACC fusions in GBM, the FGFR3-TACC3 cDN A was cloned from OSC-1 123 and recombinant lentiviruses were prepared expressing FGFR3-TACC3, FGFR1 -TACC1 , a kinase-dead FGFR3-TACC3 protein (FGFR3-TACC3-K508M), wild type FGFR3 and wild type TACC3. Transduction of RatlA fibroblasts and Jnk4A;Arf-/~ astrocytes with the FGFR3-TACC3 lentivirus resulted in the expression of the fusion protein at levels comparable to those present in GSC-1 Ϊ 23 (Figure 11), Having reconstituted in non- transformed cells the endogenous level of the FGFR-TACC protein that accumulates in GBM cells, it was determined whether it was sufficient to initiate oncogenic transformation in vitro and in vivo. Ratl cells expressing FGFR3-TACC3 and FGFRl-TACCl but not those expressing FGFR3-TACC3-K508M, FGFR3, TACC3 or the empty lentivirus acquired the ability to grow in anchorage-independent conditions in soft agar (Figure 3A). Transduction of the same lentiviruses in primary Ink4A;Arf-/- astrocytes followed by subcutaneous injection into immunodeficient mice revealed that only astrocytes expressing FGFR3-TACC3 and FGFR l -TACC l formed tumors. The tumors emerged in 100% of the mice injected with astrocytes expressing the fission proteins and were glioma-like lesions with strong positivity for K167, phospho-histone H3, nestin, GFAP and 01ig2 (Figure 3B).
[60269] Next, it was determined whether the FGFR3-TACC3 fusion protein is oncogenic when transduced to a small number of ceils directly into the brain of immunocompetent animals. A recently described mouse glioma model was used in which brain tumors are initiated by lentiviral transduction of oncogenes and inactivation of p53 in the mouse brain (Marumoto et ai, 2009), To target adult NSCs, the adult mouse hippocampus was stereotactically transduced with purified lentivirus expressing the FGFR3-TACC3 protein and shRNA against p53 (pTomo-FGFR3-TACC3-shp53). Seven of eight mice (87.5%) transduced with FGFR3--TACC3 succumbed from malignant brain tumors within 240 day s (Figure 3C). None of the mice transduced with a lentivirus expressing the most frequent gain-of- function mutation in GBM (the constitutively active EGFRvTTI, pTomo-EGFRviil- shp53 ) or the pTomo-shp53 control lentivirus died or developed clinical signs of brain tumors (Figure 3C). The FGFR3-TACC3 tumors were high-grade glioma with strong propensity to invade the normal brain and stained positive for the glioma stem cell markers nestin and Qlig2 and the glial marker GFAP. They were also highly positive for Ki67 and phospho- histone H3, thus displaying rapid tumor growth (Figure 3D), The expression of FGFR3- TACC3 in the xenograft and intracranial tumor models was comparable to the expression of the endogenous protein in the human GSCs and tumor (Figure 11D, HE and 11F).
[00270] These data show that FGFR -TACC fusion proteins possess transforming activity in two independent cellular models and this acti vity is not the result of the overexpression of individual FGF R and TACC genes. They also show that direct transduction of the FGFR3- TACC3 protein to the adult mouse brain leads to efficient development of malignant glioma.
[00271] The FGFR-TACC fusions interfere with mitotic progression and induce chromosome missegregation and aneuploidy. To elucidate the mechanism by which the FGFR-TACC fusion drives oncogenesis, it was explored whether it activates downstream FGFR signaling. FGFR3-TACC3 failed to hyperactivate the canonical signaling events downstream of FGFR (pERK and pAKT) in the presence or a bsence of the ligands FGF-1 , FGF-2 or FGF-8 (Wesche et al., 201 1) (Figures 12 A, 12B and 12C). However, FGFR3- TACC3 displayed constitutive phosphorylation of its TK domain and the adaptor protein FRS2, both of which were abolished by the specific inhibitor of FGFR-associated TK activity PD 173074 (Mohammad! et al, 1998) or the K508M mutation (Figure 4.4). Thus, FGFR3- TACC3 gains constitutive kinase activity that is essential for oncogenic transformation but the downstream signaling of this aberrant acti vity is distinct from the canonical signaling events downstream to FGFR. By driving the localization of the fusion protein, the TACC domain can create entirely novel TK-dependent functions. The TACC domain is essential for the localization of TACC proteins to the mitotic spindle (Hood and Royie, 201 1 ; Peset and Vemos, 2008). Confocal imaging showed that FGFR3-TACC3 designed an arc-shaped structure bending over and encasing the metaphase spindle poles, frequently displaying asymmetry towards one of the two poles and relocated to the nridbody as cells progressed into the late stages of mitosis (telophase and cytokinesis) (Figure 4B and 12D). Conversely, the localization of TACC3 was restricted to spindle microtubules and did not reiocalize to the midbody (Figure 12E). Wild ty e FGFR3 lacked discrete localization patterns in mitosis (Figure 12F).
[00272] The mitotic localization of FGFR3-TACC3 indicates that it may impact the fidelity of mitosis and perturb the accurate delivery of the diploid chromosomal content to daughter cells, thus generating aneuploidy. Mitotic progression of individual cells was examined in vector-transduced and FGFR3-TACC3 expressing cells co-expressing histone H2B-GFP by time-lapse microscopy. The average time from nuclear envelope breakdown to anaphase onset was increased in cells expressing FGFR3-TACC3 in comparison with control cells. The mitotic delay was further exacerbated by difficulties in completing cytokinesis (Figures 4C and 4D).
[60273] Next, it was determined whether the expressions of the FGFR-TACC fusion proteins induce defects of chromosomal segregation. Quantitative analyses of mitoses revealed that ceils expressing FGFR3-TACC3 or FGFR1-TACC1 exhibit a three to five fold increase of chromosomal segregation errors than control cells. The most frequent mitotic aberrations triggered by the fusion proteins were misaligned chromosomes during metaphase, lagging chromosomes at anaphase and chromosome bridges that impaired cytokinesis and generated micronuclei in the daughter cells (Figures 4E, 4F and 13A). Aberrations at the metaphase-anaphase transition frequently lead to the inability of mitotic cells to maintain a metaphase arrest after treatment with a spindle poison. Over 18% of cells expressing FGFR3-TACC3 displayed prematurely separated sister chromatids in contrast with less than 3% in control, FGFR3 or TACC3-expressing cells (Figures 13B and 13C). Accordingly, cells expressing the fusion protein were unable to efficiently arrest in metaphase after nocodazole treatment (Figure 13D),
[Θ0274] The above findings indicate that expression of the FGFR3-TACC3 fusion protein may spark aneuploidy. Karyotype analysis revealed that FGFR3-TACC3 increased over 2.5 fold the percent of aneuploidy and led to the accumulation of cells with broad distribution of chromosome counts in comparison with cells transduced with empty vector, FGFR3 or TACC3 (Figure 5A). Accordingly, GSC-l 123 contained aneuploid modal number of chromosomes (49) and manifested a broad distribution of chromosome counts characterized by 60% of metaphase spreads that deviate from the mode (Table 7).
G027S] Table 7: Chromosome analysis by SKY of 2Θ cells from the GSC-1123 culture
[00276] Next, it was determined whether aneuploidv is a direct consequence of FGFR3- TACC3 expression and is induced in human diploid neural cells. Primary human astrocytes analyzed six days after transduction with the FGFR3-TACC3 lentivirus exhibited a 5 -fold increase of the rate of aneupJoidy and a significantly wider distribution of chromosome counts (Figures SB, 5C and SD). Consistent that aneuploidv is detrimental to cellular fitness, acute expression of FGFR3-TACC3 compromised the proliferation capacity of human astrocytes. However, continuous culture of FGFR3-TACC3-expressing human astrocytes led to progressive gain of proliferative capacity that overrode that of control cells (Figure 4.4, 14B). Thus, the acute expression of FGFR3-TACC3 in primary normal hitman cells from the central nervous system causes CIN and aneuploidy with an acute fitness cost manifested by slower proliferation.
[00277] It was also determined whether the CIN and aneuploidy caused by FGFR3- TACC3 requires the TK activity of FGFR3 and can be corrected. Treatment with PD 173074 rescued the aneuploidy caused by FGFR3-TACC3 by over 80%, restored the narrow distribution of chromosome counts typical of control cells and largely corrected the cohesion defect (Figures 6A, 6B and 6C). Together, these findings indicate that the CIN and aneuploidy caused by rearrangements of FGFR and TACC genes are reversible and suggest that specific FGFR kinase inhibition may be a valuable therapeutic strategy in tumor cells expressing FGFR-TACC fusion proteins.
[00278] FGFR-TACC fusion proteins are new therapeutic targets in GEM. Driver genetic alterations trigger a state of oncogene addiction in the cancer cells harboring them that can be exploited therapeutically. To ask whether FGFR-TACC fusions confer addiction to FGFR-TK activity, cell growth was analyzed in the presence of PD 173074, AZD4547 or BGJ398, the latter being two highly specific inhibitors of FGFR-TK under clinical investigation (Gavine et al., 2012; Guagnano et al., 201 1). Each of the three drugs inhibited growth of ceils expressing FGFR3-TACC3 and FGFR1-TACC1 at concentrations <10 nM whereas they were ineffective at concentrations as high as 1 μΜ in cells transduced with vector, FGFR3, TACC3 and the FGFR3-TACC3-K508M mutant (Figures 7A, 14C and 14D), These findings underscore the ele vated degree of specificity for FGFR kinase inhibition towards cells carrying the fusion protein. The growth of GSC-1 12.3 cells, which naturally harbor the FGFR3-TACC3 translocation, was also abolished by nanomolar concentrations of FGFR-TK inhibi tors (Figure 7B). Targeting of the fusion gene by FGFR3 shR A inhibited the growth of cells ectopicaily expressing FGFR3-TACC3 and GSC- 1 123 proportionally to the silencing efficiency of FGFR3-TACC3 (Figures 7C and 14E).
[0(5279] Finally, it was determined whether treatment with PD173074 of mice bearing glioma xenografts of FGFR3-TACC3 transformed astrocytes inhibits tumor growth. Twelve days after injection of tumor cells, subcutaneous tumors were present in all animals. The mice were randomized in two cohorts and treated with PD173074 or vehicle. PD173074 elicited a potent growth inhibition of FGFR3-TACC3 glioma (Figure 71>). To confirm the efficacy of a clinically meaningful FGFR-TK inhibitor using a more anatomically relevant model, the AZD4547 FGFR inhibitor, a compound under clinical investigation (Gavine et al., 2012), was used against intracranial luciferase-expressing FGFR3-TACC3 -driven glioma xenografts. After an engraftment period, tumor-bearing animals were treated with either AZD4547 or vehicle. Oral administration of AZD4547 markedly prolonged survival (Figure 7E). Taken together, the data provide a strong rationale for a clinical trial based on FGFR inhibitors in GBM harboring FGFR -TACC rearrangements.
[00280] DISCUSSION
[00281] This work has established that recurrent, oncogenic and addicting gene fusions identify a subset of GBM patients. The functional characterization of FGFR-TACC fusions indicates that the constitutively active FGFR-TK and the TACC domain of the fusion protein are both essential for oncogenesis. The TACC- dependent mis-localization to mitotic cells of the FGFR kinase results in aberrant compartmentaiization of a constitutively active TK to the mitotic spindle pole, thus providing a mechanistic explanation for the impaired mitotic fidelity, chromosome mis-segregation and aneuploidy instigated by the fusion protein.
[$0282] Without being bound by theory, mutation of the genes that con trol chromosome segregation during mitosis can explain the high rate of CIN and aneuploidy, which is typical of most solid tumors including GBM (Gordon et al., 2012). A few examples of mutational inactivation of candidate genes have been reported in human cancer (Solomon et al., 201 1 ; Thompson et al., 2010). However, gain-of- function mutations causally implicated in the control of mitotic fidelity have not been described. This clashes with the classical observ ation from cell fusion experiments that the underlying mechanisms that cause CIN behave as dominant traits, indicating that the CIN phenotype results from gain-of-function events rather than gene inactivation (Lengauer et al., 1997, 1998). The FGFR-TACC gene fusion is a novel mechanism for the initiation of CIN and provides a clue to the nature of dominant mutations responsible for aneuploidy in human cancer.
[00283] The rapid emergence of mitotic defects and aneuploid cell populations triggered by the fusion protein in normal human astrocytes, combined with the correction of aneuploidy after short inhibition of FGFR-TK activity indicate that aneuploidy is a key event in tumor induction by the FGFR-TACC gene fusions. Induction of aneuploidy per se is detrimental to cellular fitness (Sheltzer and Anion, 201 1). Full-blown tumorigenesis requires cooperation between aneuploidy and genetic lesions that confer growth advantage and protect cells against the detrimental effects of aneuploidy (Coschi and Dick, 2012; Holland and Cleveland, 2009; Weaver and Cleveland, 2009). Therefore, the potent tumor-initiating activity of FGFR-TACC fissions shows that the novel oncoproteins have growth-promoting signaling functions that complement the loss of mitotic fidelit with ensuing karyotypic alterations (Sheltzer and Amon, 201 1).
[00284] Targeted therapies against common genetic alterations in GBM have not changed the dismal clinical outcome of the disease, most likely because they have systematically failed to eradicate the truly addicting oncoprotein activities of GBM. The highly specific antitumor effects and the correction of aneupioidy precipitated by FGFR-TK inhibition of FGFR- TACC-driven GBM provide a strong rationale for clinical trials based on FGFR inhibitors in patients harboring FGFR-TACC rearrangements. The computational gene fusion discovery pipeline reported here detected other GBM cases in which FGFR family genes are implicated in additional gene fusions beyond the FGFR-TACC rearrangements. Therefore, the frequency of 3.1 % is likely to be an underestimate of the target GBM patient population that may benefit from FGFR-TK inhibition.
[00285] EXPERIMENTAL PROCEDURES
[00286] Cell culture and isolation and maintenance of GSCs. Ra i A, mouse astrocytes Ink4A;Arf-/-, and human astrocytes were cultured in DMEM supplemented with 10% PBS. Isolation and culture of GSCs was performed as described (Cairo et al., 2010). For treatment in vitro with PD 173074, AZD4547 or BJG398, ceils infected with vector control, FGFR3, TACC3, FGFR-TACC fusions or FGFR3 -TA CC3 -K508M were seeded in 96-well plates and treated with increasing concentrations of FGFR inhibitors. After 72-120 h, growth rate was measured using the 3-(4,5-dimethythiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. Data were expressed as mean±SD. Proliferation rate in GSC-1123 infected with FGFR3 shRNA lentivims was determined by plating dissociated gliomaspheres at 2x104 cells/well in twelve-well plates 5 days after infection. The number of viable cells was determined by trypan blue exclusion in triplicate cultures obtained from triplicate independent infections. Cell number was scored every other day.
[00287] BNA, RNA preparation, genomic and real-time quantitative PCR (qRT-PCR).
[0(5288] The validation of fusion transcripts was performed rising both genomic and RT- PCR with forward and reverse primer combinations designed within the margins of the paired-end read sequences detected by RNA-seq. DNA, RNA preparation and qRT-PCR were performed as described (Carro et al, 2.010; Zhao et al., 2008). To identify novel fusion transcripts within the GBM cohort, PGR primers pairs were designed to bind upstream to the TK domain of the FGFR genes and inside or downstream the Coiled Coil domain of the T'ACC genes. Expressed fusion transcript variants were subjected to direct sequencing to confirm sequence and frame. Primer sequences are included below.
{0028.9] Subcutaneous xenografts and drug treatment. Rati A or Ink4A;Arf-/- astrocytes (Sx l O5) transduced with different lentiviral constructs were suspended in 150 μί of PBS, together with 30 μΐ of Matrigel (BD Biosciences), and injected subcutaneously in the flank of athymic nude (Nu/Nu) mice (Charles River Laboratories, Wilmington, MA). For experiments with FGFR inhibitors, mice carrying -200-300 mm"5 subcutaneous tumors derived from Ink4A;Arf-/- astrocytes were randomized to receive 50 mg/kg PD 173074 in 0.05 M lactate buffer (pH 5) or an equal volume of lactate buffer by oral gavage. Treatment was administered for three cycles consisting of four consecutive days followed by two days of rest. Tumor diameters were measured with caliper, and tumor volumes estimated using the formula: 0.5 x length x width7'. Data are expressed as mean±SE. Mice were sacrificed whenumors in the control group reached the maximal size allowed.
[1)029(5] Orthotopic transplantation and drug treatment. Ink4A;Arf~/- astrocytes carrying a luciferase expressing vector were transduced with FGFR3-TACC3 ientivitus, 1x10'' ceils in 2 μΐ of saline were injected in the caudate-putamen of 4-6 week old male athymic nude (Nu/Nu) mice using a stereotaxic frame (coordinates relative to bregma: 0.5 mm anterior; 1.1 mm lateral; 3.0 mm ventral) and a 26 gauge Hamilton syringe. Six days after injection, mice underwent bioluminescence imaging using a Xenogen CCD apparatus and were randomized to receive 50 mg kg AZD4547 in 1% Tween 80 (treatment group) or DMSO in an equal volume of vehicle by oral gavage (con trol group). AZD4547 was administered daily for two cycles of 10 days with a two day interval. Mice were monitored daily and sacrificed when neurological symptoms appeared. Kaplan-Meier survival curve was generated using the DNA Statview software package (AbacusConcepts, Berkeley, CA). Log-rank analysis was performed on the Kaplan-Meier survival curve to determine statistical significance.
[0(5291] intracranial injecthms of lentiviruses. Intracranial injection of FGFR3-TACC3- shp53, or shp53 pTomo lentiviruses was performed in 4 week-old
C57/BL/6J mice in accordance with guidelines of 1ACUC Committee. Briefly, 1.8 μΐ of purified lentiviral particles in PBS (lX109/ml) were injected into the dentate gyms using a stereotaxic frame (coordinates relative to bregma: 1.45 mm posterior; 1 ,65 mm lateral; 2.4 mm ventral) and a 26 gauge Hamilton syringe. Mice were monitored daily and sacrificed when neurological symptoms appeared. Mouse brain was analyzed histopafhologically and by immunofluorescence staining.
[00292] Histology and i munostaining. Tissue preparation and immunohistochemistry on brain tumors and immunofluorescence staining were performed as previously described (Carro et al., 2010; Zhao et al., 2009; Zhao et al., 2008). Antibodies used in immunostaining and immuno blotting are listed below.
[00293] Clo ing and Lentiviral production, Lentivirus preparation and infections were performed as described (Carro el al., 2010) and are detailed in Extended Experimental Procedures.
[80294] Karyotype analysis. Cultured cells were coicemid (2.0 ng/ml) treated for 90 minutes before harvesting for karyotopic analysis as detailed in Extended Experimental procedures. At least one hundred cells in metaphase were examined for chromosome count. PMSCS was scored in cells where a majority of the sister chromosomes were no longer associated. Two-tailed unpaired t-tcsts with Welch's correction were performed for comparison of means analysis.
[00295] Immunofluorescence and live-cell microscopy. Immunofluorescence microscopy was performed on ceils fixed with 4% PFA in PHEM (60 mM Pipes, 27 mM Hepes, 10 mM EGTA, 4 mM MgS04, pH 7.0). Cells were permeabilized using 1% Triton X-100. Mitotic spindles were visualized by anti-a-tubulin antibody (Sigma). Secondary antibodies conjugated to Alexa Fluor-488/-594 (Molecular Probes) were used. Ail staining with multiple antibodies were performed in a sequential manner. DN A was stained by DAPI (Sigma). Fluorescence microscopy was performed on a Nikon AIR MP microscope.
[00296] Identification of gene fusions from whole transcriptome (RNA-seq) and exome sequencing. RNA- Sequencing was performed from total RNA extracted from GSC cultures isolated from nine GBM patients using lllumina HiSeq 2000, producing roughly 60.3 million paired reads per sample. Using the global alignment software Burrows- Wheeler Aligner (BWA) (Li and Durbin, 2009) with modified Mott's trimming, an initial seed length of 32, maximum edit distance of 2 and a maximum gap number of 1, on average 43.1 million reads were mapped properly to the RefSeq transcriptome and, of the remaining, 8.6 million were mapped to the hg!9 genome per sample. The remaining 14.3% of paired reads— including those that failed to map to either transcriptome or genome with proper forward-reverse (F-R) orientation, within expected insert size, and with minimal soft clipping (unmapped portions at the ends of a read)— were considered to be appropriate for gene fusion analysis,
[00297] A novel computational pipeline was constructed called ΐΧ-F e that identifies two sources of evidence for the presence of a gene fusion: 1 . Split inserts, in which each read of a mate pair maps entirely to one side of a breakpoint, and 2. Individual split reads that span a breakpoint. Split inserts are readily detected from BWA mapping. On the other hand, split reads demand precision alignment of smaller nucleotide stretches. To that end, the pipeline employs the local alignment package BLAST with word size of 20, identity cutoff of 95%, expectation cutoff of 10"4, and soft filtering to map raw paired reads against the RefSeq transcriptome. From this procedure, a list of potential split reads were obtained that were filtered to ensure maintenance of coding frame in the predicted fission transcript given the proper F-R orientation in the read pair. False positive candidates produced from paralogous gene pairs were also screened out using the Duplicated Genes Database and the
EnsemblCompara GeneTrees (Vileila et ai, 2009). Pseudogenes in the candidate list were annotated using the list from HUGO Gene Nomenclature Committee (HGNC) database (Seal et al., 201 1) and given lower priority. For each remaining gene fission candidate, a virtual reference was created based on the predicted fusion transcript and re-mapped all unmapped reads using BLAST with word size of 16, identity cutoff of 85%, query coverage greater than 85%, and expectation cutoff of 10 to obtain a final count of split reads and inserts.
Moreover, sequencing depth per base of the virtual reference was calculated to corroborate that components of each gene participating in the gene fusion were highly expressed.
[ΘΘ298] To establish the recurrence of the initial panel of gene fusion candidates, the gene fusion discover}- pipeline was modified to produce EXome-Fme, which probes for fusions within the available dataset of paired-read exome DNA sequencing of 84 matched GBM samples from TCGA. To increase sensitivity for gene fusion identification, reads unmapped by BWA were aligned to the gene pair participating in each fusion candidate using a BLAST word size of 24 for split inserts and 16 for split read and split insert discovery. Given that the breakpoint detected in DNA cannot directly indicate the resulting breakpoint in the transcribed RNA, no restriction was made on split insert orientation. For split reads, it was only required that the component of the split read mapped to the same gene as its mate maintained F-R directionality. [0(5299] Co-outlier expression and CNV analysis from TCGA GEM samples. Tomlins et al. (Tomlins et al., 2005) reported thai outlier gene expression from mieroarray datasets identifies candidate oncogenic gene fusions. Wang et al. (Wang et al., 2009) suggested a "breakpoint principle" for intragenic copy number aberrations in fusion partners. The two principles (outlier expression and intragenic CNV) were combined to identify candidate gene fusions in GBM samples from Atlas-TCGA. Genomic and expression data sets were downloaded from TCGA public dat portal as available on December I, 2011, where a description of TCGA data types, platforms, and analyses is also available (2008). Specific data sources were (according to Data Levels and Data Types) as follows: Expression data, "Level 2" normalized signals per probe set (Affymetrix HT_HG-U 33A) of 84 samples; Copy number data, "Level 1" raw signals per probe (Affymetrix Genome- Wide Human SNP Array 6.0) of the 4 FGFR3-TACC3 gene fusion positive samples (tumor and matched normal control).
[00360] The gene expression analysis was performed first using R3. The median absolute deviation (MAD) was calculated and then a gene was labeled as an outlier according to the following formula: Zy = 0.6745(x;,.; - mean(x;) )/MADi > 3.5 Oglewicz and Roaglin, 1993). Samples were identified as ECFS (expression candidate fission sample) if both genes of interest (e. g. FGFR3 and TACC3) displayed outlier behavior (co- outliers). Next ECFS were analyzed for CNV using pennCNV (Wang et al., 2007). Tumors samples were paired to their normal controls to obtain the log ratio values and the VEGA algorithm was used to obtain a more accurate segmentation (Morganella et al, 2010).
[00301] Karyotypic Analysis. The colcemid treated cells were tiypsinized, centrifuged for 7 minutes at 200 x g, and the cell pellet re-suspended in warmed hypotonic solution and incubated at 37°C for 3 minutes. The swollen cells were then centrifuged and the pellet re- suspended in 8 mi of Carnoy's fixative (3: 1 methanol :glacial acetic acid). The cell suspension was centrifuged and washed twice in Carnoy 's fixative. After the last centrifugation, the cells were resuspended in 0.5 to 1 ml of freshly prepared fixative to produce an opalescent cell suspension. Drops of the final cell suspension were placed on clean slides and air-dried. Slides were stained with DAPI and metaphases were analyzed under a fluorescent microscope,
[00302] Cloning and Lentiviral production. Lentiviral expression vectors, pLOC-GFP (Open Biosystems) and pTomo-shp53, were used to clone FGF 3, TACC3, FGFR3-TACC3, FGFR3-TACC3-K508M, and FGFR1 -TACC1. pTorno-shp53 was a gift oflnder Venna and Dinorah Friedman-Morvinski (Salk Institute, San Diego). The FGFR3-TACC3- 508M mutant was generated using the Phusioti Site Direct Mutagenesis kit (NEB, U SA). MISSION shRNAs clones (pLKO. l lentiviral expression vectors) against FGFR3 were purchased from Sigma. The hairpin sequences targeting the FGFR3 gene are-
[00303] 5'-TGCGTCGTGGAGAACAAGTTT-3' (OTCN0000000372; Sh#2) (SEQ ID NO: 182);
[0Θ3Θ4] 5'-GTTCCACTGCAAGGTGTACAG-3 ' (#TRCN0000430673; Sh#3) (SEQ ID NO: 183);
[003115] 5'-GCACAACCTCGACTACTACAA-3' (#TRCN()()()0000374; Sh#4) (SEQ ID NO: 184).
[00306] Genomic and mR A RT-PCR. Total RNA was extracted from cells by using RNeasy Mini Kit (QIAGEN), following the manufacturer instructions. 500 ng of total RNA was retro-transcribed by using the Superscript ill kit (Invitrogen), following the manufacturer instructions. The cDNAs obtained after the retro-transcription was used as templates for qPCR. The reaction was performed with a Roche480 thermal cycler, by using the Absolute Blue QPCR SYBR Green Mix from Thermo Scientific. The relative amount of specific mRNA was normalized to 18S. Results are presented as the mean ± SD of triplicate amplifications.
[003Θ7] Primers used are: liFGFR3 -RT-FW 1 : 5'-GTAACCTGCGGGAGTTTCTG-3' (SEQ ID NO: 162); liFGFR3 -RT-RE V 1 : 5 '-ACACCAGGTCCTTGAAGGTG-3 ' (SEQ ID NO: 163); hTACC3-RT-FW2: 5 '-CCTGAGGGACAGTCCTGGTA-3 ' (SEQ ID NO: 164); hTACC3-RT-REV2: 5'-AGTGCTCCCAAGAAATCGAA-3 ' (SEQ ID NO: 165); hWRAP53-RT-FWl : 5'-AGAGGTGACCACCAATCAGC-3' (SEQ ID NO: 180); hWRAP53-RT-REVl : 5'-CGTGTCCCACACAGAGACAG-3' (SEQ ID NO: 181 ).
[00308] Primers used for the screening of FGFR-TACC fusions are: FGFR3-FW1 : 5 '-CGTGAAGATGCTGAAAG ACG ATG-3 ' (SEQ ID NO: 166); TACC3-REV 1 : 5'- AAACGCTTGAAGAGGTCGGAG-3' (SEQ ID NO: 167); FGFR1-FW1 : 5 ' - ATGCTAGC AGGGGTCTCTGA-3 ' (SEQ ID NO: 168);
TACC1 -REV1 : 5'-CCCTTCCAGAACACCTTTCA-3' (SEQ ID NO: 169).
[00309] Primers used for genomic detection of FGFR3-TACC3 fusion in GBM-1123 and GSC-1 123 are:
Genomic FGFR3-FW1 : 5' -ATGATCATGCGGG AGTGC-3 ' (SEQ ID NO: 170); genomicTACC3-REVl : 5 ' · GGGGGTCGAACTTGAGGTAT-3 ' (SEQ ID NO: 171).
[00310] Primers used to validate fusions detected by RNA-seq are:
POLR2A-FW1 : 5 '-CGCAGGCTI I GTAGTGAG-3 ' (SEQ ID NO: 172);
WRAP53-REV1 : 5 ' -TGTAG GCG CG AAAGGAAG -3 ' (SEQ ID NO: 173);
P1GU-FW1 : 5'-GAACTCATCCGGACCCCTAT-3' (SEQ ID NO: 174);
NCQA6-REV1 : 5 '-GCTTTCCCCATTGCACTTTA-3 ' (SEQ ID NO: 175);
ST8SIA4-FW1 : 5 ' -GAG GAG AGAAG CA CGTGGAG-3 ' (SEQ ID NO: 176);
PAM.-REV1 : 5'-GGCAGACGTGTGAGGTGTAA-3' (SEQ ID NO: 177);
CAPZB-FW: 5 '-GTGATCAGCAGCTGGACTGT-3 ' (SEQ ID NO: 178);
UBR4-REV1 : 5 ' -GAGCCTGGGCATGGATCT-5 ' (SEQ ID NO: 179).
[Θ0311] Coiifocai microscopy imaging. For immunofluorescence of fixed cells, images were recorded with a Z-optical spacing of 0.25 μηι using a Nikon AIR MP and a 60X1 .3 oil objective and analyzed using Image.! software (National Institute of Health). For live-cell analyses. Rati A cells infected with pLNCX-H2B retrovirus and transduced with lentiviral vector or FGFR3-TACC3 fusion were seeded in glass bottom dishes in phenol red free DMEM and followed by time-lapse microscopy using the Nikon AIR MP biostation at 37°C and 5% CQ?/95 % air. Images with a Z-optical spacing of 1 um were recorded every 4 min for 8 h. Images of unchal lenged mitosis from early prophase until cytokinesis were processed using Image! software (N ational Institute of Health). The time-point of nuclear envelope breakdown (NEB) was defined as the first frame showing loss of smooth appearance of chromatin and anaphase was the first frame when chromosome movement towards the poles became apparent. Nuclear envelope reconstitution (NER) was defined as the first frame showing nuclei decondensation,
[ΘΘ312] Box and whisker plots were calculated from image sequences from at least 50 recorded cells. Two-tailed unpaired t-tests with Welch's correction were performed for comparison of means analysis using StatView software (AbacusConcepts, Berkeley, CA).
[00313] Immunofluorescence. Antibodies and concentrations used in
immunofluorescence staining are:
Anti-Ki67 Rabbit 1 1000 Vector Labs
Anti-pH H3 Rabbit 1 500 Miliipore
Anti-FGFR3 Mouse 1 1000 Santa Cruz
Anti-Tacc3 Goat 1 1000 USBioiogicai
Anti-a-tubuiin Mouse 1 1000 Sigma
Anti-Nestln Mouse 1 1000 BD Pharmingen
Anti-Olig2 Rabbit 1 200 I BL
Anti-GFAP Rabbit 1 200 Dako
Anti-ERK Rabbit 1 1000 Ceil Signaling
Anti-pERK Rabbit 1 1000 Ceil Signaling
AntiFRS Rabbit 1 250 Santa Cruz
Anti-pFRS Rabbit 1 1000 Cell Signaling
Anti-AKT Rabbit 1 1000 Ceil Signaling
Anti-pAKT473 Rabbit 1 1000 Cell Signaling
[00314] REFERENCES
Ablain, J., Nasr, ., Bazarbachi, A., and de The, H. (201 1). The Drug-Induced Degradation of Oncoproteins: An Unexpected Achilles' Heel of Cancer Cells? Cancer Discov. 1, 1 17-127.
Bass, A.J., Lawrence, M.S., Brace, L.E., Ramos, A.H., Drier, Y., Cibuiskis, K., Sougnez, C, Voet, D., Saksena, G., Sivachenko, A., et ah (201 1). Genomic sequencing of colorectal adenocarcinomas identifies a recurrent VTT.1 -TCF7L2 fission. Nat. Genet. 43, 964-968.
Cahill, D.P., Kinzier, K.W., Vogeistein, B., and Lengauer, C. (1999). Genetic instability and darwinian selection in tumours. Trends Cell. Biol. 9, M57-60. Carro, M.S., Lim, W.K., Alvarez, M.J., Bollo, R.J., Zhao, X., Snyder, E.Y., Sulrnan, E.P., Anne, S.L., Doetseh, F., Colman, H„ el al. (2010), The transcriptional network for mesenchymal transformation of brain tumours. Nature 463, 318-325.
Coschi, C.H., and Dick, F.A. (2.012). Cliromosome instability and deregulated proliferation: an unavoidable duo. Cell Mol. Life Sci. 69, 2009-2024
Druker, B.j. (2009). Perspectives on the development of iniaiinib and the future of cancer research. Nat. Med. 15, 1 149- 1 152.
Fumari, F.B., Fenton, T., Bachoo, R.M., Mukasa, A., Stommel, J.M., Siegh, A., Hahn, W.C., Ligon, .L., Louis. D.N., Brennan, C, el al. (2007). Malignant astrocytic glioma: genetics, biology, and paths to treatment. Genes Dev. 21, 2683-2710.
Gavine, P.R., Mooney, L.. Kilgour, E., Thomas, A.P., Al-Kadhimi, ., Beck, S., Rooney, C, Coleman, T., Baker, D., Meilor, M.J., et al (2012). AZD4547: An Orally Bioavailable, Potent, and Selective inhibitor of the Fibroblast Growth Factor Receptor Tyrosine Kinase Family, Cancer Res. 72, 2045-2056.
Gerber, D.E., and Minna, J.D. (2010). ALK inhibition for non-small cell lung cancer: from discovery to therapy in record time. Cancer Cell 18, 548-551.
Gordon, D.J., Resio, B., and Pellman, D. (2012), Causes and consequences of aneuploidy in cancer. Nature reviews Genet. 13, 189-203.
Guagnano, V., Furet, P., Spanka, C, Bordas, V., Le Douget, M., Stamm, C, Brueggen, L, Jensen, M.R., Schneil, C, Schmid, H„ et al. (201 1). Discovery of 3 -(2,6-dichioro-3,5 - dimethoxy-phenyl)- 1 - {6-[4-(4-ethyl-piperazin- 1 -yl)-phenylamin ol-pyrimidin-4-yi}- l- methyl-urea (NVP-BGJ398), a potent and selective inhibitor of the fibroblast growth factor receptor family of receptor tyrosine kinase. J. Med. Chem. 54, 7066-7083.
Holland, A.J., and Cleveland, D.W. (2009). Boveri revisited: chromosomal instability, aneuploidy and tumorigenesis. Nat. Rev. Mol. Cell. Biol, 10, 478-487.
Hood, F.E., and Royle, S.J. (201 1). Pulling it together: The mitotic function of TACC3. Bioarchitecture 1, 105- 109. Lee, J., Kotliarova, S., Kotliarov, Y., Li, A., Su, Q., Donin, N.M., Pastorino, S., Purow, B.W., Christopher, N., Zhang, W., et ai. (2006), Tumor stem cells derived from glioblastomas cultured in b'FGF and EGF more closely mirror the phenotype and genotype of primary tumors than do serum-cultured cell lines. Cancer Cell 9, 391-403.
Lengauer, C, Kinzler, K.W., and Vogelstein, B. ( 1997). Genetic instability in colorectal cancers. Nature 386, 623-627.
Lengauer, C, Kinzler, K.W., and Vogelstein, B. (1998). Genetic instabilities in human cancers. Nature 396, 643-649.
Lo, H.W. (2010). EGFR-targeted therapy in malignant glioma: novel aspects and mechanisms of drug resistance. Curr. Moi. Pharmacol. 3, 37-52.
Marumoto, T., Tashiro, A., Friedmann-Morvinski, D., Scadeng, M., Soda, Y., Gage, F.H., and Verma, i.M. (2009). Development of a novel mouse glioma model using lentiviral vectors. Nat. Med. 15, 1 10- 1 16.
Mitelman, F., Johansson, B., and Mertens, F. (2007). The impact of translocations and gene fusions on cancer causation. Nat. Rev. Cancer 7, 233-245.
Mohammadi, M., Fro m, S., Hamby, J.M., Schroeder, M.C., Panek, R.L., Lu, G.H., Eliseenkova, A.V., Green, D., Schlessinger, J., and Hubbard, S.R. (1998). Crystal structure of an angiogenesis inhibitor bound to the FGF receptor tyrosine kinase domain. EMBO J. 17, 5896-5904.
Qhgaki, H., and Kleihues, P. (2005). Population-based studies on incidence, survival rates, and genetic alterations in astrocytic and oligodendroglial gliomas. J. Neuropathol. Exp. Neurol. 64, 479-489.
Peset, 1., and Vernos, 1. (2008). The TACC proteins: TACC-ling microtubule dynamics and centrosome function. Trends Cell. Biol. 18, 379-388.
Prensner, J.R., and Chinnaiyan, A.M. (2009). Oncogenic gene fusions in epithelial carcinomas. Curr Opin Genet. Dev. 19, 82-91.
Reardon, D.A., Desjardins, A., Vredenburgh, J.J., Gumrangan, S., Friedman, A.H., Herndon, IE., 2nd, Marcelio, I, Norileet, J.A., McLendon, R.E., Sampson, J.H., et al (2010). Phase 2 trial of erlotinib plus siro limits in adults with recurrent glioblastoma. J. Neurooncol. 96, 219-
230.
Sheltzer, J.M., and Anion, A. (2011). The aneupioidy paradox: costs and benefits of an incorrect karyotype. Trends Genet. 27, 446-453.
Solomon, D.A., Kim, T., Diaz-Martinez, L.A., Fair, J., Elkahioun, A.G., Harris, B.T., Toretsky, J. A., Rosenberg, S.A., Shukla, N., Ladanyi, M., et al. (201 1). Mutational inactivation of STAG2 causes aneupioidy in human cancer. Science 333, 1 39-1043.
Stephens, P.J., McB ide, D.J., Lin, M.L., Varela, L, Pleasance, E. ., Simpson, J.T., Stebbings, L.A., Leroy, C, Edkins, S., Mudie, L.J., et al. (2009). Complex landscapes of somatic rearrangement in human breast cancer genomes. Nature 462, 1005- 1010.
Still, I.H., Vince, P., and Cowell, J.K. (1999). The third member of the transforming acidic coiled coil-containing gene family, TACC3, maps in 4 i6, close to translocation breakpoints in multiple myeloma, and is upregulated in various cancer cell lines. Genomics 58, 165-170.
Thompson, S.L., Bakhoum, S.F., and Compton, D.A. (2010). Mechanisms of chromosomal instability. Curr. Biol. 20, R285-295.
TomJins, S.A., Laxman, B., Dhanasekaran, S.M., Heigeson, B.E., Cao, X., Morris, D.S., Menon, A., ling, X., Cao, Q., Han, B,, et al, (2007). Distinct classes of chromosomal rearrangements create oncogenic ETS gene fissions in prostate cancer. Nature 448, 595-599.
Tomlins, S.A., Rhodes, D.R., Perner, S., Dhanasekaran, S.M., Mehra, R., Sun, X.W., Varambaily, S., Cao, X., Tchinda, J., Kuefer, R., et al (2005), Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science 310, 644-648.
Turner, N., and Grose, R. (2010), Fibroblast growth factor signalling: from development to cancer. Nat. Rev. Cancer 10, 1 16-129. van den Bent, M.J., Brandes, A. A., Rampling, R., Kouwenhoven, M.C., Kros, J.M., Carpentier, A.F., Clement, P.M., Frenay, M., Campone, M., Baurain, J.F., et al, (2009). Randomized phase II trial of erlotinib versus temozoiomide or carmastine in recurrent glioblastoma: EORTC brain tumor group study 26034. J. Clin. Oncol. 27, 1268-1274. Wang, X.S., Prensner, J.R., Chen, G., Cao, Q., Han, B., Dhanasekaran, S.M., Ponnala, R., Cao, X., Varambally, S,, Thomas, D.G., et al. (2009). An integrative approach to reveal driver gene fissions from paired-end sequencing data in cancer. Nat. Biotechnol. 27, 1005- 101 1 .
Weaver, B.A., and Cleveland, D.W. (2009). The role of aneuploidy in promoting and suppressing tumors. J. Cell. Biol. 185, 935-937.
Wescbe, J., Plaglund, K., and Haugsten, E.M. (201 1). Fibroblast growth factors and their receptors in cancer. Biochem. J. 437, 199-213.
Yan, PL, Parsons, D.W., Jin, G., McLendon, R., Rasheed, B.A., Yuan, W., Kos, L, Batinie- Haberle, L, Jones, S., Riggins, G.J., el al. (2.009). 1DH1 and IDH2 mutations in gliomas. New Engl J. Med. 360, 765-773.
Zhao, X., D, D.A., Lim, W.K., Brahmaehary, M., Carro, M.S., Ludwig, T., Cardo, C.C., Guillemot, F., Aldape, K., Califano, A„ et al, (2009). The N-Myc-DLL3 cascade is suppressed by the ubiquitin ligase Pluwei to inhibit proliferation and promote neurogenesis in the developing brain. Dev. Cell 17, 210-221.
Zhao, X., Heng, J.L, Guardavaecaro, D., Jiang, R., Pagano, M., Guillemot, F., lavarone, A., and Lasorella, A, (2008). The HECT-domain ubiquitin ligase Huwel controls neural differentiation and proliferation by destabilizing the N-Myc oncoprotein. Nat Cell Biol 10, 643-653.
(2008). Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455, 1061 -1068. iglewicz, B., and Hoaglin, D.C. (1993). Flow to detect and handle outliers (Milwaukee, Wis.: ASQC).
Li, H., and Durbin, R. (2009). Fast and accurate short read alignment with Burrows-Wheeler transform, Bioinformatics 25, 1754- 1760.
Morganella, S., Cerulo, L., Viglietto, G., and Ceccarelli, M. (2010). VEGA: variational segmentation for copy number detection. Bioinformatics 26, 3020-3027. Seal, R.L., Gordon, S.M., Lush, M.I, Wright, M.W., and Bruford, E.A. (201 1 ). genenames.org: the HGNC resources in 201 1. Nucleic Acids Res 39, D514-519.
Tomlins, S.A., Rhodes, D.R., Perner, S., Dhanasekaran, S.M., Mehra, R., Sun, X.W., Varambally, S., Cao, X., Tchinda, J., Kuefer, R., et al. (2005). Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science 310, 644-648.
Vilella, A.J., Severin, J., Ureta-Vidal, A., Heng, L., urbin, R., and Birney, E. (2009). EnsemblCompara GeneTrees: Complete, duplication-aware phylogenetic trees in vertebrates. Genome Res. 19, 327-335.
Wang, K., Li, M., Hadley, D., Liu, R., Giessner, J.. Grant, S.F., Hakonarson, H., and Bucan, M. (2007). PennCNV: an integrated hidden Markov model designed for high-resolution copy number variation detection in whole-genome SNP gcnotyping data. Genome Res. 1 7, 1665- 1674.
Wang, X.S., Prensner, J.R., Chen, G., Cao, Q., Han, B., Dhanasekaran, S.M., Ponnala, R., Cao, X., Varambally, S., Thomas, D.G., et al. (2009). An integrative approach to reveal driver gene fusions from paired-end sequencing data in cancer. Nat. Biotechnol. 27, 1005- 101 1.
[ΘΘ315] Example 2- Fusions in GBM
n.ci.: not done
- i - [00317] Table 9: Subcutaneous tumor xenografts
n.d : not dene
[0(5318] Ta e j): Analysis of chromosomal number in Rati cells
[00319] Table 11 : Analysis of chromosomal number in Iranian astrocytes
[$$320] Example 3 - Fusions in Other Cancers
[00321] The inventors previously reported in Example i thai 3.1 % of human glioblastoma harbor FGFR3-TACC3 and FGFRl -TACCl gene fusions. Tumors harboring FGFR3- TACC3 gene fusions are identified by the presence of highly specific focal micro- amplification events of the rearranged portions of the FGFR3 and T ACC3 genes (See FIG. 2E). Therefore, these micro-amplification events can be used as distinctive marks for the presence of FGFR3-TACC3 gene fusions. It was asked whether other types of human tumors also harbor FGFR3-TACC3 gene fusions from the analysis of Copy Number Variations (CNVs) of SNP arrays generated from the Atlas-TCGA project. This analysis was performed using segmented CNVs data visualized using the ntegrated Genomic Viewers software. The analysis revealed that the following tumors, shown in the FIGS. 31-35, display focal micro- amplification events of FGFR3 and TACC3 that indicate the presence of FGFR3-TACC3 gene fusions (in FIGS. 31-3S, red indicates amplification (A), blue indicates deletion (D); FIG. 31: Bladder Urothelial Carcinoma; FIG. 32: Breast Carcinoma; FiG. 33: Colorectal Carcinoma; FIG. 34: Lung Squamous Cell Carcinoma; FIG. 35: Head and Neck Squamous Ceil Carcinoma).
[00322] Taken together, these data indicate that the same FGFR3-TACC3 gene fusions reported for (he first time in Glioblastoma also occur in several other types of human tumors. Therefore, as for Glioblastoma and other epithelial cancers (such as the human tumors discussed herein), the identification of FGFR-TACC gene fusions also provides a new diagnostic and therapeutic target for treatment with drugs that inhibit FGFR-TACC gene fusions.

Claims (66)

What is claimed Is:
1 . A purified fusion protein comprising a tyrosine kinase domain of an FGFR protein fused to a polypeptide that constitutiv eiy activates the tyrosine kinase domain of the FGFR protein.
2. A purified fusion protein comprising a transforming acidic coiled-coil (TACC)
domain fused to a polypeptide with a tyrosine kinase domain, wherein the TACC domain constitutiveiy activates the tyrosine kinase domain.
3. A purified fusion protein comprising the tyrosine kinase domain of an FGFR protein fused 5' to the TACC domain of a transforming acidic coiied-coi!-containing (TACC) protein.
4. The purified fusion protein of claim 1 or 3, wherein the FGFR protein is FGFRI , FGFR2, FGFR3, or FGR4.
5. The purified fusion protein of claim 3, wherein the TACC protein is TACC1 , TACC2, 01- TACC3.
6. A purified fusion protein encoded by an FGFRI -TACC 1 nucleic acid, wherein
FGFRI -TACC 1 comprises a combination of exons 1-17 of FGFRI located on human chromosome 8pl 1 spliced 5' to a combination of exons 7- 13 of TACC 1 located on human chromosome 8pl 1 , wherein a genomic breakpoint occurs in any one of exons 1-17 of FGFRI and any one of exons 7-13 of TACC 1.
7. A purified fusion protein encoded by an FGFR2-TACC2 nucleic acid, wherein
FGFR2-TACC2 comprises a combination of any exons 1 -18 of FGFR2 located on human chromosome K)q26 spliced 5" to a combination of any exons 1-23 of TACC2 located on human chromosome K)q26.
8. A purified fusion protein encoded by an FGFR3-TACC3 nucleic acid, wherein FGFR3-TACC3 comprises a combination of exons 1-16 of FGFR3 located on human chromosome 4 l6 spliced 5' to a combination of exons 8- 16 of TACC3 located on hitman chromosome 4p 16, wherein a genomic breakpoint occurs in any one of exons 1 -16 of FOFR3 and any one of exons 8-16 of TACC3.
9. A purified fusion protein encoded by an FGFR3-TACC3 nucleic acid, wherein
FGFR3-TACC-3 comprises a combination of exons 1 -16 of FGFR3 located on human chromosome 4pl6 spliced 5' to a combination of exons 8-16 of TACC3 located on human chromosome 4 l6, wherein a genomic breakpoint occurs in any one oiinirons 1-16 of FGFR3 and any one of exons 8-16 of TACC3.
10. A purified fusion protein encoded by an FGFR3-TACC3 nucleic acid, wherein
FGFR3-TACC3 comprises a combination of exons 1-16 of FGFR3 located on human chromosome 4 l 6 spliced 5' to a combination of exons 8-16 of TACC3 located on human chromosome 4 l6, wherein a genomic breakpoint occurs in any one of exons 1-16 of FGFR3 and any one of introm 7-16 of TACC3.
1 1. A purified fusion protein encoded by an FGFR3-TACC3 nucleic acid, wherein
FGFR3-TACC3 comprises a combination of exons 1 -16 of FGFR3 located on human chromosome 4 l6 spliced 5' to a combination of exons 8-16 of TACC3 located on human chromosome 4p 16, wherein a genomic breakpoint occurs in any one of introns 1-16 of FGFR3 and any one of Introm 7-16 of TACC3.
12. A synthetic nucleic acid encoding the fusion protein of claim 1 , 2, 3, 6, 7, 8, 9, 10, or 1 1.
13. A purified FGFR3-TACC3 fusion protein comprising SEQ ID NO: 79, 158, 159, 160, or 161.
14. A purified FGFR3-TACC3 fusion protein having genomic breakpoint comprising at least 3 consecutive amino acids from amino acids 730-758 of SEQ ID NO: 90 and comprising at least 3 consecutive amino acids from amino acids 549-838 of SEQ ID NO: 92.
15. A purified FGFR3-TACC3 fusion protein having a genomic breakpoint comprising SEQ ID NO: 78.
16. A purified FGFR3-TACC3 fusion protein having a genomic breakpoint comprising any one of SEQ ID NOS: 85, 86, 87, or 89.
17. A purified FGFRl -TACCl fusion protein comprising SEQ ID NO: 150.
1 8. A purified FGFRl -TACCl fusion protein having a genomic breakpoint comprising at least 3 consecutive amino acids from amino acids 746-762 of SEQ ID NO: 146 and comprising at least 3 consecutive amino acids from amino acids 572-590 of SEQ ID NO: 148.
19. A purified FGFRl -TACC 1 fusion protein having a genomic breakpoint comprising SEQ ID NO: 88.
20. A synthetic nucleic acid encoding an FGFR3-TACC3 fusion protein comprising SEQ ID NO: 94.
21. A synthetic nucleic acid encoding an FGFR3-TACC3 fusion protein having a
genomic breakpoint comprising at least 9 consecutive in-frame nucleotides from nucleotides 2443-2530 of SEQ ID NO: 91 and comprising at least 9 consecutive in- frame nucleotides from nucleotides 1800-2847 of SEQ ID NO: 93.
22. A synthetic nucleic acid encoding an FGFR3-TACC3 fusion protein having a genomic breakpoint comprising any one of SEQ ID NOS: 1 -77.
23. A synthetic nucleic acid encoding an FGFRl-TACCi fusion protein comprising SEQ I D NO: 1 51.
24. A synthetic nucleic acid encoding an FGFRl-TACCi fusion protein having a
genomic breakpoint comprising at least 9 consecutive in-frame nucleotides from nucleotides 3178-3228 of SEQ ID NO: 147 and comprising at least 9 consecutive in- frame nucleotides from nucleotides 2092-2794 of SEQ ID NO: 149.
25. A synthetic nucleic acid encoding an FGFRl -TACCi fusion protein having a
genomic breakpoint comprising SEQ ID NO: 83.
26. An antibody or antigen-binding fragment thereof, that specifically binds to a purified fusion protein comprising a tyrosine kinase domain of an FGFR protein fused to a polypeptide that constitutively activates the tyrosine kinase domain of the FGFR protein.
27. 'T'he antibody or antigen-binding fragment of claim 26, wherein the FGFR protein is FGFR1 , FGFR2, FGFR3, or FGFR4.
28. 'T'he antibody or antigen-binding fragment of claim 26, wherein the fusion protein is an FGFR-TACC fusion protein.
29. 'T'he antibody or antigen-binding fragment of claim 28, wherein the FGFR-TACC fusion protein is FGFRl -TACC i , FGFR2-TACC2, or FGFR3-TACC3.
30. T'he antibody or antigen-binding fragment of claim 29, wherein the FGFRl -TACC i fusion protein comprises the amino acid sequence of SEQ ID NO: 150.
31. The an tibody or antigen-binding fragment of claim 29, wherein the FGFR3-TACC3 fusion protein comprises the amino acid sequence of SEQ ID NO: 79, 158, 159, 160, or 161.
32. A composition for decreasing in a subject the expression level or activity of a fusion protein comprising the tyrosine kinase domain of an FGFR protein fused to a polypeptide that constitutively activates the tyrosine kinase domain of the FGFR protein, the composition in an admixture of a pharmaceutically acceptable carrier comprising an inhibitor of the fusion protein,
33. The composition of claim 32, wherein the fusion protem is an FGFR-TACC fusion protein.
34. The composition of claim 32, wherein the inhibitor comprises an antibody that specifically binds to a FGFR-TACC fusion protein or a fragment thereof; a small molecule that specifically binds to a FGFR protein; a small molecule that specifically binds to a TACC protein; an antisense RN A or antisense DNA that decreases expression of a FGFR-TACC fusion polypeptide; a siRNA that specifically targets a FGFR-TACC fusion gene; or a combination thereof.
35. The composition of claim 32, wherein the FGFR protein is FGFR ϊ , FGFR2, FGFR3, or FGFR4.
36. The composition of claim 33, wherein the FGFR-TACC fusion protein is FGFR 1- TACC1, FGFR2-TACC2, or FGFR3-TACC3.
37. The composition of claim 34, wherein the small molecule that specifically binds to a FGFR protein comprises AZD4547, NVP-BGJ398, PD173074, NF449, TK1258, BIBF-1 120, BMS-582664, AZD-2171 , TSU68, AB1010, AP24534, E-7080, LY2874455, or a combination thereof.
38. A method for decreasing in a subject in need thereof the expression level or activity of a fusion protein comprising the tyrosine kinase domain of an FGFR protein fused to a polypeptide that constitutive!}' activates the tyrosine kinase domain of the FGFR protein, the method comprising:
(a) administering to the subject a therapeutic amount of a composition of claim 32; and
(b) determining whether the fusion protein expression level or activity is decreased compared to fusion protein expression level or activity prior to administration of the composition, thereby decreasing the expression le vel or activity of the fusion protein.
39. A method for treating a gene-fusion associated cancer in a subject in need thereof, the method comprising administering to the subject an effective amount of a FGFR fusion rnoiecuie inhibitor.
40. A method of decreasing growth of a solid tumor in a subject in need thereof, the method comprising administering to the subject an effective amount of a FGFR fusion molecule inhibitor, wherein the inhibitor decreases the size of the solid tumor.
41. The method of claim 39, wherein the gene-fusion associated cancer comprises
glioblastoma multiforme, breast cancer, lung cancer, prostate cancer, or colorectal carcinoma.
42. The method of claim 40, wherein the solid tumor comprises glioblastoma multiforme, breast cancer, lung cancer, prostate cancer, or colorectal carcinoma.
43. The method of claim 39 or 40, wherein the FGFR fusion protein comprises an FGFR protein fused to a polypeptide that constitutively activates the tyrosine kinase domain of the FGFR protein.
44. The method of claim 38, 39 or 40, wherein the fusion protein is an FGFR-TACC fusion protein.
45. The method of claim 39 or 40, wherein the inhibitor comprises an antibody that
specifically binds to a FGFR-TACC fusion protein or a fragment thereof: a small molecule that specifically binds to a FGFR protein; a small molecule that specifically binds to a TACC protein; an antisense RNA or antisense DNA that decreases expression of a FGFR-TACC fusion polypeptide; a siRNA that specifically targets a FGFR-TACC fusion gene; or a combination thereof.
46. The method of claim 38 or 43, wherein the FGFR protein is FGFR] , FGFR2, FGFR3, or FGFR4.
47. The method of claim 45, wherem the FGFR-TACC fusion protein is FGFRl-TACCL FGFR2-TACC2, or FGFR3-TACC3.
48. The method of claim 45, wherem the small molecule that specifically binds to a
FGFR protein comprises AZD4547, NVP-BGJ398, PD 173074, NF449, TK1258, BIBF-1 120, BMS-582664, AZD-2171, TSU68, ABiOl O, AP24534, E-7080, LY2874455, or a combination thereof.
49. A diagnostic kit for determining whether a sample from a subject exhibits a presence of a FGFR fusion, the kit comprising at least one oligonucleotide that specifically hybridizes to a FGFR fusion, or a portion thereof.
50. The kit of claim 49. wherein the oligonucleotides comprise a set of nucleic acid primers or in situ hybridization probes.
51. The kit of claim 49. wherein the oligonucleotide comprises SEQ ID NO: 162, 163, 164,165, 166, 167, 168, 169, or a combination thereof.
52. The kit of claim 50, wherein the primers prime a polymerase reaction only when a FGFR fusion is present,
53. The kit of claim 49, wherein the determining comprises gene sequencing, selective hybr dization, selective amplification, gene expression analysis, or a combination thereof,
54. A diagnostic kit for determining whether a sample from a subject exhibits a presence of a FGFR fusion protein, the kit comprising an antibody that specifically binds to a FGFR fusion protein comprising SEQ ID NO: 79, 85, 86, 87, 88, 89, 150, 158, 159, 160, or 161 , wherein the antibody will recognize the protein only when a FGFR fusion protein is present.
55. The kit of claim 54, wherein the antibody directed to and FGFR fusion comprising SEQ ID NO: 79, 85, 86, 87, 88, 89, 150, 158, 159, 160, or 161.
56. The kit of claim 49 or 54, wherein the FGF -fusion is an FGFR-TACC fusion.
57. The kit of claim 49 or 54, wherein the FGFR is FGFRl, FGFR2, FGFR3, or FGFR4.
58. The kit of claim 55, wherein the FGFR-TACC fusion is FGFRl -TACC1, FGFR2- TACC2, or FGFR3-TACC3.
59. A method for detecting the presence of a FGFR fusion in a hitman subject, the method comprising:
(a) obtaining a biological sample from the human subject; and
(b) detecting whether or not there is a FGFR fusion present in the subject,
60. 'T'he method of claim 59, wherein the detecting comprises measuring FGFR fusion protein levels by ELISA using an antibody directed to SEQ ID NO: 79, 85, 86, 87, 88, 89, 150, 158, 159, 160, or 161 ; western blot using an antibody directed to SEQ ID NO: 79, 85, 86, 87, 88, 89, 150, 158, 159, 160, or 161; mass spectroscopy, isoelectric focusing, or a combination thereof.
61. A method for detecting the presence of a FGFR fusion in a human subject, the method comprising:
(a) obtaining a biological sample from a human subject; and
(b) detecting whether or not there is a nucleic acid sequence encoding a FGFR fusion protein in the subject.
62. The method of claim 61 , wherein the nucleic acid sequence comprises any one of SEQ TD NOS: 1-77, 80-84, or 95-145.
63. The method of claim 61, wherein the detecting comprises using hybridization,
amplification, or sequencing techniques to detect a FGFR fusion.
64. The method of claim 63, wherein the amplification uses primers comprising SEQ ID NO: 162, 163, 164, 165, 166, 167, 168, or 169.
65. The method of claim 59 or 61 , wherein the FGFR-fusion is an FGFR-TACC fusion.
66. The method of claim 59 or 61 , wherein the FGFR is FGF 1 , FGFR2 , FGFR3, or FGFR4.
The method of claim 65, wherein the FGFR-TACC fusion is FGFRl-TACC l , FGFR2-TACC2, or FGFR3-TACC3.
A method of initiating oncogenic transformation in vitro, (he method comprising:
(a) transducing cells cultured in vitro with FGFR-TACC fusion DNA; and
(b) determining whether the cells acquire the ability to grow in anchorage- independent conditions, form multi-layered foci, or a combination thereof,
A method of initiating oncogenic transformation in vivo, the method comprising:
(a) transducing cells cultured in vitro with FGFR-TACC fusion DN A;
(b) injecting a mouse with the transduced cells; and
(c) detennining whether a. tumor grows in the mouse.
The method of claim 69, wherein the injecting is a subcutaneous or intracranial injection.
A method of identifying a compound that decreases the oncogenic activity of a FGFR-TACC fusion, the meihod comprising:
(a) transducing a cell cultured in vitro with FGFR-TACC DNA;
(b) contacting a cell with a !igand source for an effective period of time; and
(c) determining whether the cells acquire the ability to grow in anchorage- independent conditions, form multi-layered foci, or a combination thereof, compared to cells cultured in the absence of the test compound.
AU2013295805A 2012-07-24 2013-07-24 Fusion proteins and methods thereof Active AU2013295805B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201261675006P 2012-07-24 2012-07-24
US61/675,006 2012-07-24
PCT/US2013/051888 WO2014018673A2 (en) 2012-07-24 2013-07-24 Fusion proteins and methods thereof

Publications (2)

Publication Number Publication Date
AU2013295805A1 true AU2013295805A1 (en) 2015-02-12
AU2013295805B2 AU2013295805B2 (en) 2019-05-02

Family

ID=49997966

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2013295805A Active AU2013295805B2 (en) 2012-07-24 2013-07-24 Fusion proteins and methods thereof

Country Status (8)

Country Link
EP (2) EP2877854B1 (en)
JP (1) JP6382194B2 (en)
KR (1) KR102132149B1 (en)
CN (1) CN104797936B (en)
AU (1) AU2013295805B2 (en)
CA (1) CA2880013C (en)
ES (1) ES2932954T3 (en)
WO (1) WO2014018673A2 (en)

Families Citing this family (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130079241A1 (en) 2011-09-15 2013-03-28 Jianhua Luo Methods for Diagnosing Prostate Cancer and Predicting Prostate Cancer Relapse
WO2013059740A1 (en) 2011-10-21 2013-04-25 Foundation Medicine, Inc. Novel alk and ntrk1 fusion molecules and uses thereof
JP6107812B2 (en) 2012-03-08 2017-04-05 アステラス製薬株式会社 Novel FGFR3 fusion
EP2877854B1 (en) 2012-07-24 2022-09-07 The Trustees of Columbia University in the City of New York Fgfr-tacc fusion proteins and methods thereof
US20150203589A1 (en) 2012-07-24 2015-07-23 The Trustees Of Columbia University In The City Of New York Fusion proteins and methods thereof
TWI606066B (en) 2012-09-27 2017-11-21 中外製藥股份有限公司 FGFR3 Fusion Gene and Its Targeted Medicine
EP2914621B1 (en) 2012-11-05 2023-06-07 Foundation Medicine, Inc. Novel ntrk1 fusion molecules and uses thereof
AU2013337264B2 (en) * 2012-11-05 2018-03-08 Foundation Medicine, Inc. Novel fusion molecules and uses thereof
WO2014111744A1 (en) 2013-01-16 2014-07-24 INSERM (Institut National de la Santé et de la Recherche Médicale) A soluble fibroblast growth factor receptor 3 (fgr3) polypeptide for use in the prevention or treatment of skeletal growth retardation disorders
WO2014113729A2 (en) 2013-01-18 2014-07-24 Foundation Mecicine, Inc. Methods of treating cholangiocarcinoma
JP2016515508A (en) 2013-03-15 2016-05-30 ザ・トラスティーズ・オブ・コロンビア・ユニバーシティ・イン・ザ・シティ・オブ・ニューヨーク Fusion protein and method thereof
CA2935216C (en) 2013-12-30 2021-11-09 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Fusion genes associated with progressive prostate cancer
CN106458999B (en) 2014-03-26 2019-12-03 阿斯特克斯治疗有限公司 Combination
SG10201808565QA (en) * 2014-03-31 2018-11-29 Debiopharm Int Sa Fgfr fusions
EP3193944B1 (en) 2014-07-17 2021-04-07 University of Pittsburgh - Of the Commonwealth System of Higher Education Methods of treating cells containing fusion genes
EP3198033B1 (en) * 2014-09-26 2022-02-16 Janssen Pharmaceutica NV Use of fgfr mutant gene panels in identifying cancer patients that will be responsive to treatment with an fgfr inhibitor
WO2016105517A1 (en) * 2014-12-23 2016-06-30 The Trustees Of Columbia University In The City Of New York Fusion proteins and methods thereof
JOP20200201A1 (en) 2015-02-10 2017-06-16 Astex Therapeutics Ltd Pharmaceutical compositions comprising n-(3,5-dimethoxyphenyl)-n'-(1-methylethyl)-n-[3-(1-methyl-1h-pyrazol-4-yl)quinoxalin-6-yl]ethane-1,2-diamine
US10478494B2 (en) 2015-04-03 2019-11-19 Astex Therapeutics Ltd FGFR/PD-1 combination therapy for the treatment of cancer
WO2017027473A1 (en) 2015-08-07 2017-02-16 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Methods for predicting prostate cancer relapse
EP3693459A1 (en) * 2015-10-10 2020-08-12 Guardant Health, Inc. Methods and applications of gene fusion detection in cell-free dna analysis
KR20220122762A (en) 2016-07-07 2022-09-02 화이자 인코포레이티드 Soluble fibroblast growth factor receptor 3 (SFGFR3) polypeptides and uses thereof

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5229501A (en) * 1991-01-11 1993-07-20 Chiron Corporation Expression and use of human fibroblast growth factor receptor
WO1999035159A1 (en) * 1998-01-08 1999-07-15 Brigham & Women's Hospital, Inc. Lymphoma/leukemia oncogene, oncoprotein and methods of use
US7868149B2 (en) * 1999-07-20 2011-01-11 Monsanto Technology Llc Plant genome sequence and uses thereof
US20060275794A1 (en) * 2005-03-07 2006-12-07 Invitrogen Corporation Collections of matched biological reagents and methods for identifying matched reagents
JP2007021008A (en) * 2005-07-20 2007-02-01 Hitachi Ltd Magnetic resonance imaging device equipped with dnp hyperpolarization means
ES2366160T3 (en) * 2006-11-28 2011-10-17 Aventis Pharma S.A. FC FUSIONS WITH RECEIVER FOR FGF SOLUBLE MODIFIED WITH IMPROVED BIOLOGICAL ACTIVITY.
WO2008070179A2 (en) * 2006-12-06 2008-06-12 Monsanto Technology, Llc Genes and uses for plant improvement
EP2165715B1 (en) * 2007-05-29 2013-12-18 Sapporo Medical University Therapeutic agent for cancer, and method for treatment of cancer
EP2171452A4 (en) * 2007-06-22 2010-10-20 Expression Pathology Method of discovering and analyzing secreted biomarkers of disease from solid tissue
EP2080812A1 (en) * 2008-01-18 2009-07-22 Transmedi SA Compositions and methods of detecting post-stop peptides
CN104788564A (en) * 2009-03-25 2015-07-22 健泰科生物技术公司 Anti-FGFR3 antibodies and methods using same
US20110195848A1 (en) * 2010-01-08 2011-08-11 Roopra Avtar S Gene expression and breast cancer
US8945847B2 (en) * 2010-05-24 2015-02-03 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Methods and kits for ascertaining biosafety of an agent
CN102219860B (en) * 2011-05-20 2012-09-12 烟台荣昌生物工程有限公司 FGFR-Fc fusion protein and applications thereof
AU2013227139B2 (en) * 2012-02-28 2017-02-16 Astellas Pharma Inc. Nitrogen-containing aromatic heterocyclic compound
JP6107812B2 (en) * 2012-03-08 2017-04-05 アステラス製薬株式会社 Novel FGFR3 fusion
EP2877854B1 (en) 2012-07-24 2022-09-07 The Trustees of Columbia University in the City of New York Fgfr-tacc fusion proteins and methods thereof

Similar Documents

Publication Publication Date Title
US11920138B2 (en) Fusion proteins and methods thereof
AU2013295805B2 (en) Fusion proteins and methods thereof
AU2013295805A1 (en) Fusion proteins and methods thereof
US20230303985A1 (en) Fusion proteins and methods thereof
US10023855B2 (en) Fusion protein comprising C-terminal domain of RET protein and use thereof as a diagnosing marker
Hwang et al. Insulin-like growth factor-II mRNA binding protein-3 and podoplanin expression are associated with bone invasion and prognosis in oral squamous cell carcinoma
KR20140098834A (en) Erbb3 mutations in cancer
WO2015048367A1 (en) Fusion proteins and methods of use thereof
EP3778644A2 (en) Fgfr-tacc fusion proteins and methods thereof
EP2894169B1 (en) Fusion protein comprising axl and composition for treating cancer comprising same
CN111575241B (en) T lymphocyte of chimeric chondroitin sulfate proteoglycan 4 receptor and preparation method and application thereof
WO2020138503A1 (en) Therapeutic and prophylactic agent for glioma, brain tumor malignancy marker, brain tumor prognostic marker, method for determining malignancy and prognosis of brain tumor and antibody inhibiting tumor proliferation
O’Connell et al. WNT2B Deficiency Causes Enhanced Susceptibility to Colitis Due to Increased Inflammatory Cytokine Production
Lian Characterization of isoform specific RET knockdown in cancer cell lines
Bencivenga et al. Role of Twist1 transcription factor in thyroid tumor progression
Macpherson A study of p120-catenin and its tyrosine phopshorylation in cancer cell adhesion and invasion
Boon The HGF/MET pathway in colorectal cancer