Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
INCRNAS FOR THERAPY AND DIAGNOSIS OF ANGIOGENESIS
Document Type and Number:
WIPO Patent Application WO/2015/162172
Kind Code:
A2
Abstract:
The present invention relates to a pharmaceutical composition comprising (i) a compound promoting the expression and/or the activity of one or more long non-coding RNAs (IncRNAs) selected from SEQ ID NOs 1 to 22, preferably selected from SEQ ID NOs 1 to 3; and/or (ii) a compound inhibiting the expression and/or the activity of one or more IncRNAs selected from SEQ ID NOs 23 to 42. The present invention also relates to a pharmaceutical composition comprising (i) a compound promoting the expression and/or the activity of one or more IncRNAs selected from SEQ ID NOs 23 to 42; and/or (ii) a compound inhibiting the expression and/or the activity of one or more long non-coding RNAs (IncRNAs) selected from SEQ ID NOs 1 to 22, preferably selected from SEQ ID NOs 1 to 3.

Inventors:
THUM THOMAS (DE)
FIEDLER JAN (DE)
Application Number:
PCT/EP2015/058709
Publication Date:
October 29, 2015
Filing Date:
April 22, 2015
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
HANNOVER MED HOCHSCHULE (DE)
International Classes:
C12N15/113
Attorney, Agent or Firm:
VOSSIUS & PARTNER (Siebertstraße 3, München, DE)
Download PDF:
Claims:
CLAIMS

A pharmaceutical composition comprising

(i) a compound promoting the expression and/or the activity of one or more long non-coding RNAs (IncRNAs) selected from SEQ ID NOs 1 to 22, preferably selected from SEQ ID NOs 1 to 3; and/or

(ii) a compound inhibiting the expression and/or the activity of one or more IncRNAs selected from SEQ ID NOs 23 to 42.

A compound (i) promoting the expression and/or the activity of one or more IncRNAs selected from SEQ ID NOs 1 to 22, preferably selected from SEQ ID NOs 1 to 3; and/or (ii) inhibiting the expression and/or the activity of one or more IncRNAs selected from SEQ ID NOs 23 to 42 for use in treating or preventing ischemia, preferably cardiac ischemia and most preferably coronary artery disease, or for use in promoting wound healing.

The pharmaceutical composition of claim 1 or compound for use according to claim 2, wherein the compound as defined in (i) is

(a) a nucleic acid sequence which comprises or consists of the nucleic acid sequence of one or more IncRNAs selected from SEQ ID NOs 1 to 22 or an nucleic acid sequence which is at least 70% identical thereto,

(b) an expression vector expressing the nucleic acid sequence as defined in (a), preferably under the control of a heart-specific promoter, or

(c) an host comprising the expression vector of (b).

The pharmaceutical composition of claim 1 or compound for use according to claim 2, wherein the compound as defined in (i) is

(a) a transcription factor promoting the expression of one or more IncRNAs selected from SEQ ID NOs 1 to 22, and/or

(b) a small molecule enhancing the expression of one or more IncRNAs selected from SEQ ID NOs 1 to 22.

The pharmaceutical composition or compound for use according to any one of claims 1 to 4, wherein the compound as defined in (ii) is

(a) a nucleic acid sequence which comprises or consists of a nucleotide sequence being complementary to at least 12 continuous nucleotides of a IncRNAs selected from SEQ ID NOs 23 to 42,

(b) a nucleic acid sequence which comprises or consists of a nucleotide sequence which is at least 70% identical to the complementary strand of one or more IncRNAs selected from SEQ ID NOs 23 to 42,

(c) a nucleic acid sequence which comprises or consists of a nucleotide sequence according to (a) or (b), wherein U is replaced by T,

(d) an expression vector expressing the nucleic acid sequence as defined in any one of (a) to (c), preferably under the control of a heart-specific promoter, or

(e) an host comprising the expression vector of (d).

The pharmaceutical composition or compound for use according to any one of claims 1 to 4, wherein the compound as defined in (ii) is an aptamer, a ribozyme, an antibody, a protein drug, or a small molecule inhibitor.

A pharmaceutical composition comprising

(i) a compound promoting the expression and/or the activity of one or more IncRNAs selected from SEQ ID NOs 23 to 42; and/or

(ii) a compound inhibiting the expression and/or the activity of one or more long non- coding RNAs (IncRNAs) selected from SEQ ID NOs 1 to 22, preferably selected from SEQ ID NOs 1 to 3.

A compound (i) promoting the expression and/or the activity of one or more IncRNAs selected from SEQ ID NOs 23 to 42; and/or

(ii) inhibiting the expression and/or the activity of one or more IncRNAs selected from SEQ ID NOs 1 to 22, preferably selected from SEQ ID NOs 1 to 3

for use in treating or preventing a tumor, preferably a hypoxic tumor, or for use in treating or preventing diabetic retinopathy.

The pharmaceutical composition of claim 7 or compound for use according to claim 8, wherein the compound as defined in (i) is

(a) a nucleic acid sequence which comprises or consists of the nucleic acid sequence of one or more IncRNAs selected from SEQ ID NOs 23 to 42or an nucleic acid sequence which is at least 70% identical thereto,

(b) an expression vector expressing the nucleic acid sequence as defined in (a), preferably under the control of a heart-specific promoter, or

(c) an host comprising the expression vector of (b). The pharmaceutical composition of claim 7 or compound for use according to claim 8, wherein the compound as defined in (i) is

(a) a transcription factor promoting the expression of one or more IncRNAs selected from SEQ ID NOs 23 to 42, and/or

(b) a small molecule enhancing the expression of one or more IncRNAs selected from SEQ ID NOs 23 to 42.

The pharmaceutical composition or compound for use according to any one of claims 7 to 10, wherein the compound as defined in (ii) is

(a) a nucleic acid sequence which comprises or consists of a nucleotide sequence being complementary to at least 12 continuous nucleotides of a IncRNAs selected from SEQ ID NOs 1 to 22,

(b) a nucleic acid sequence which comprises or consists of a nucleotide sequence which is at least 70% identical to the complementary strand of one or more IncRNAs selected from SEQ ID NOs 1 to 22,

(c) a nucleic acid sequence which comprises or consists of a nucleotide sequence according to (a) or (b), wherein U is replaced by T,

(d) an expression vector expressing the nucleic acid sequence as defined in any one of (a) to (c), preferably under the control of a heart-specific promoter, or

(e) an host comprising the expression vector of (d).

The pharmaceutical composition or compound for use according to any one of claims 7 to 10, wherein the compound as defined in (ii) is an aptamer, a ribozyme, an antibody, a protein drug, or a small molecule inhibitor.

A method for diagnosing hypoxia in a patient, comprising

(a) detecting the expression level of one or more IncRNAs selected from SEQ ID NOs 1 to 42, preferably selected from SEQ ID NOs 1 to 3 in a sample obtained from said patient; and

(b) comparing said expression level of the one or more IncRNAs with the expression level of these one or more IncRNAs in a sample obtained from healthy subjects, wherein

a greater than 2-fold upregulation of one or more IncRNAs selected from SEQ ID NOs 1 to 22; and/or

a greater than 2-fold downregulation of one or more IncRNAs selected from SEQ ID NOs 23 to 42

is indicative for hypoxia in the patient.

14. A Kit for diagnosing hypoxia in a patient, said kit comprising means for the detection of the expression level of one or more IncRNAs selected from SEQ ID NOs 1 to 42, preferably selected from SEQ ID NOs 1 to 3 and instructions how to use the kit.

15. The pharmaceutical composition, compound for use, method or kit of any one of claims 1 to 14, wherein the one or more IncRNAs are at least 3 IncRNAs, and preferably at least 5 IncRNAs.

Description:
IncRNAs for therapy and diagnosis of angiogenesis

The present invention relates to a pharmaceutical composition comprising (i) a compound promoting the expression and/or the activity of one or more long non-coding RNAs (IncRNAs) selected from SEQ ID NOs 1 to 22, preferably selected from SEQ ID NOs 1 to 3 and 14; and/or (ii) a compound inhibiting the expression and/or the activity of one or more IncRNAs selected from SEQ ID NOs 23 to 42. The present invention also relates to a pharmaceutical composition comprising (i) a compound promoting the expression and/or the activity of one or more IncRNAs selected from SEQ ID NOs 23 to 42; and/or (ii) a compound inhibiting the expression and/or the activity of one or more long non-coding RNAs (IncRNAs) selected from SEQ ID NOs 1 to 22, preferably selected from SEQ ID NOs 1 to 3 and 14.

In this specification, a number of documents including patent applications and manufacturer's manuals is cited. The disclosure of these documents, while not considered relevant for the patentability of this invention, is herewith incorporated by reference in its entirety. More specifically, all referenced documents are incorporated by reference to the same extent as if each individual document was specifically and individually indicated to be incorporated by reference. The era of non-coding RNA (ncRNA) research has been set after publishing the encyclopedia of DNA elements (ENCODE) in 2013. In fact, the majority of the genome is non-coding. The different classes of non-coding RNA comprise small microRNA (miR) and long non-coding RNA (IncRNA). To date, great effort has been taken to understand microRNA-dependent molecular mechanisms and to develop innovative therapeutic strategies. For IncRNA biology, however, very little is known. Next to well-studied miRs, IncRNAs are now in focus of different scientific disciplines.

In cancer research, the IncRNA HOTAIR has been characterized to be an oncogenic factor (Nakagawa et al., Biochem Biophys Res Comm, 2013; Kim et al., Oncogene, 2013). For example during lung cancer, HOTAIR is upregulated in patients (Nakagawa et al., Biochem Biophys Res Comm, 2013). Furthermore, asscociation of HOTAIR with Polycomb Repressor Complex 2 (PRC2) also drives pancreatic tumor expansion (Kim et al., Oncogene, 2013). In contrast, loss of XIST triggers blood cancer indicating that XIST potently inhibits cellular malformation (Yildirim et al., Cell, 2013). At a more mechanistical view, IncRNA-dependent regulation of translation has been demonstrated in breast cancer setting (Gumireddy et al., EMBO J, 2013). The role of IncRNA Fendrr has recently been discovered and highlights the importance of IncRNA abundancy during cardiac development (Grote et al., Dev Cell, 2013; Grote and Herrmann RNA Biol, 2013). Loss of Fendrr also causes embryonal death emphasizing the crucial regulatory capacity of IncRNA. Another IncRNA termed "Braveheart" is going along with this observation (Klattenhoff et al., Cell, 2013). This is an additional activator for cardiac lineage commitment where mechanistic aspects have been deciphered. Angiotensin II (Ang ll)-induced expression of IncRNA has also been investigated (Leung et al., Circ Res, 2013). Interestingly, loss of a single IncRNA reduced proliferative potential of smooth muscle cells (SMCs) and indicated participation of IncRNA for SMC-associated disease. Of great interest, non-coding ANRIL and coding gene CDKN2B could be linked in a longer study investigating leukocyte and platelet content in heart failure patients (Johnson et al., Circulation 2013).

However, the role of IncRNAs in angiogenic signalling is yet vastly unexplored. Angiogenesis is a central physiological process that establishes blood supply and oxygen supply to tissues, thereby enabling the growth and maintenance of nascent bodily structures. During angiogenesis, either pro- or anti-angiogenic signalling guides endothelial cells to sustain vascular integrity. Angiogenesis is necessary for wound healing, as well as recovery from ischemic insults. In such cases, it is beneficial to promote angiogenesis (Bahatia (2013), Mechanical and Chemical Signaling in Angiogenesis, Studies in Mechanobiology, Tissue Engineering and Biomaterials; 12(261-278)). Endothelial cell (EC) performance is of utmost importance for angiogenic signalling especially after hypoxic intervention (i.e. ischemia), e.g. myocardial infarction (Ml). So far, to the best knowledge of the inventors nothing is known for the role of specific IncRNAs in ECs. On the other hand, angiogenesis is undesirable and pathological in the context of cancerous tumors, as well as diabetic retinopathy. In these cases, it is preferable to inhibit angiogenesis (Bahatia (2013), Mechanical and Chemical Signaling in Angiogenesis, Studies in Mechanobiology, Tissue Engineering and Biomaterials; 12(261-278)). Anti-angiogenic therapy is an establsihed anti-cancer strategy that targets new blood vessels that grow to provide oxygen and nutrients to actively proliferating tumor cells (Kubota (2012); Keio J Med.; 61(2):47-56). Currently, the most established approach for influencing angiogenesis is targeting the vascular endothelial growth factor (VEGF) pathway. As is evident from the above, there is, though, an ongoing need for further therapeutic pro-angiogenic and anti-angiogenic approaches. This need is addressed by the present invention. It was surprisingly found that specific IncRNAs are involved in angiogenic signalling and therefore can be used in novel therapeutic pro-angiogenic and anti-angiogenic approaches.

Accordingly, the present invention relates in a first aspect to a pharmaceutical composition comprising (i) a compound promoting the expression and/or the activity of one or more long non-coding RNAs (IncRNAs) selected from SEQ ID NOs 1 to 22, preferably selected from SEQ ID NOs 1 to 3 and 14; and/or (ii) a compound inhibiting the expression and/or the activity of one or more IncRNAs selected from SEQ ID NOs 23 to 42.

In accordance with the present invention, the term "pharmaceutical composition" relates to a composition for administration to a patient, preferably a human patient. The pharmaceutical composition of the invention comprises the compounds recited above. It may, optionally, comprise further molecules capable of altering the characteristics of the compounds of the invention thereby, for example, stabilizing, modulating and/or activating their function. The composition may be in solid, liquid or gaseous form and may be, inter alia, in the form of (a) powder(s), (a) tablet(s), (a) solution(s) or (an) aerosol(s). The pharmaceutical composition of the present invention may, optionally and additionally, comprise a pharmaceutically acceptable carrier or excipient. Examples of suitable pharmaceutical carriers and excipients are well known in the art and include phosphate buffered saline solutions, water, emulsions, such as oil/water emulsions, various types of wetting agents, sterile solutions, organic solvents including DMSO etc. Compositions comprising such carriers or excipients can be formulated by well known conventional methods. These pharmaceutical compositions can be administered to the subject at a suitable dose. The dosage regimen will be determined by the attending physician and clinical factors. As is well known in the medical arts, dosages for any one patient depends upon many factors, including the patient's size, body surface area, age, the particular compound to be administered, sex, time and route of administration, general health, and other drugs being administered concurrently. The therapeutically effective amount for a given situation will readily be determined by routine experimentation and is within the skills and judgement of the ordinary clinician or physician. Generally, the regimen as a regular administration of the pharmaceutical composition should be in the range of 1 pg to 5 g units per day. However, a more preferred dosage might be in the range of 0.01 mg to 100 mg, even more preferably 0.01 mg to 50 mg and most preferably 0.01 mg to 10 mg per day.

Furthermore, if for example said compound is an nucleic acid sequence, such as an siRNA, the total pharmaceutically effective amount of pharmaceutical composition administered will typically be less than about 75 mg per kg of body weight, such as for example less than about 70, 60, 50, 40, 30, 20, 10, 5, 2, 1, 0.5, 0.1 , 0.05, 0.01 , 0.005, 0.001 , or 0.0005 mg per kg of body weight. More preferably, the amount will be less than 2000 nmol of nucleic acid sequence (e.g., about 4.4 x 1016 copies) per kg of body weight, such as for example less than 1500, 750, 300, 150, 75, 15, 7.5, 1.5, 0.75, 0.15, 0.075, 0.015, 0.0075, 0.0015, 0.00075 or 0.00015 nmol of iRNA agent per kg of body weight.

The length of treatment needed to observe changes and the interval following treatment for responses to occur vary depending on the desired effect. The particular amounts may be determined by conventional tests which are well known to the person skilled in the art.

Pharmaceutical compositions of the invention preferably comprise a pharmaceutically acceptable carrier or excipient. By "pharmaceutically acceptable carrier or excipient" is meant a non-toxic solid, semisolid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type (see also Handbook of Pharmaceutical Excipients 6ed. 2010, Published by the Pharmaceutical Press). The pharmaceutical composition may be administered, for example, orally, parenterally, such as subcutaneously, intravenously, intramuscularly, intraperitoneally, intrathecally, transdermal^, transmucosally, subdurally, locally or topically via iontopheresis, sublingually, by inhalation spray, aerosol or rectally and the like in dosage unit formulations optionally comprising conventional pharmaceutically acceptable carriers or excipients.

The term "ncRNA" or "non-coding RNA" as used herein designates a functional RNA molecule that is not translated into a protein. The DNA sequence from which a non-coding RNA is transcribed is often called in the art an RNA gene. The term "IncRNA" or "long non-coding RNA" is commonly used in the art and designates an ncRNA comprising more than 200 nucleotides. SEQ ID NOs 1 to 22 and 23 to 42 comprise sequences ranging from 349 to 2517 nucleotides.

The compounds of the invention may be formulated as vesicles, such as liposomes. Liposomes have attracted great interest because of their specificity and the duration of action they offer from the standpoint of drug delivery. Liposomal delivery systems have been used to effectively deliver nucleic acids, such as siRNA in vivo into cells (Zimmermann et al. (2006) Nature, 441 :1 11-114). Liposomes are unilamellar or multilamellar vesicles which have a membrane formed from a lipophilic material and an aqueous interior. The aqueous portion contains the composition to be delivered. Cationic liposomes possess the advantage of being able to fuse to the cell wall. Non-cationic liposomes, although not able to fuse as efficiently with the cell wall, are phagocytosed by macrophages and other cells in vivo. The composition of the first aspect of the invention is a pro-angiogenic pharmaceutical composition.

A compound promoting the expression of one or more IncRNAs selected from SEQ ID NOs 1 to 22 - as defined herein in item (i) - may be any compound enhancing or upregulating the transcription of an IncRNA selected from SEQ ID NOs 1 to 22. Non-limiting examples of such compounds are transcription factors enhancing the transcription of the genes encoding the IncRNAs selected from SEQ ID NOs 1 to 22 or a small molecule enhancing the expression of one or more IncRNAs selected from SEQ ID NOs 1 to 22. A transcription factor is a protein binding to specific DNA sequences, thereby controlling the transcription of genetic information from DNA to RNA. Transcription factors which enhance the expression of pro-angiogenic genes are known. A preferred example is HIF1a (hypoxia-inducible factor 1 ). Another regulatory role could be attributed to chromatin modifiers, e.g. modifiers that regulate the methylation status in CpG islands next to IncRNA genes. A small molecule is a low molecular weight compound which is by definition not a polymer. A compound promoting the activity of one or more IncRNAs selected from SEQ ID NOs 1 to 22 - as defined herein in item (i) - may be any compound which causes that said IncRNA effectively performs its function in a cell. Hence, in the simplest form such a compound may be a recombinantly produced or isolated IncRNAs selected from SEQ ID NOs 1 to 22 or any precursor or fragment thereof. In this embodiment the administration of a recombinantly produced or isolated IncRNA increases the concentration of IncRNA in the subject to be treated. This higher concentration promotes the overall activity of the respective IncRNA in the subject. The fragments have to retain or essentially retain the function of the full-length IncRNA. Such a compound may also be a vector or host being capable of producing such an IncRNAs. Hence, the fragments have to be functional fragments. Also orthologous or homologous sequences of the IncRNA selected from SEQ ID NOs 1 to 22 from different species including precursors or functional fragments thereof may be used. Alternatively, such a compound may be a compound maintaining or even enhancing the activity of an IncRNA selected from SEQ ID NOs 1 to 22 by either directly or indirectly interacting with the IncRNA. For instance, such a compound may prevent an IncRNAs selected from SEQ ID NOs 1 to 22 from degeneration by RNases or may be an interaction partner, such as another IncRNA, which binds to and promotes the activity of an IncRNA selected from SEQ ID NOs 1 to 22. Compounds as defined herein in item (i) will be further detailed herein below. The efficiency of a compound as defined herein in item (i) can also be quantified by methods comparing the level of expression and/or activity of an IncRNA selected from SEQ ID NOs 1 to 22 in the presence of a compound promoting the activity and/or expression of the IncRNA, such as a transcription factor, to that in the absence of said compound. For example, as an activity measure may be used: the change in amount of IncRNA formed. The method is preferably effected in high-throughput format as further detailed herein below.

A compound inhibiting the expression of one or more IncRNAs selected from SEQ ID NOs 23 to 42 - as defined herein in item (ii) - is in accordance with the present invention a compound lowering or preventing the transcription of one or more of the genes encoding the IncRNAs selected of SEQ ID NOs 23 to 42. Such compounds include compounds interfering with the transcriptional machinery and/or its interaction with the promoter of said genes and/or with expression control elements remote from the promoter such as enhancers. The compound inhibiting the expression of an IncRNA selected from SEQ ID NOs 23 to 42 specifically inhibits the expression of said IncRNA, for example, by specifically interfering with the promoter region controlling the expression of the IncRNA. Preferably, the transcription of an IncRNA selected from SEQ ID NOs 23 to 42 is reduced by at least 50%, more preferred at least 75% such as at least 90% or 95%, even more preferred at least 98% and most preferred by about 100%. A compound inhibiting the activity of an IncRNAs selected from SEQ ID NOs 23 to 42 - as defined herein in item (ii) - in accordance with the present invention causes said IncRNA to perform its function with lowered efficiency. The compound inhibiting the activity of an IncRNAs selected from SEQ ID NOs 23 to 42 specifically inhibits the activity of said IncRNA. Preferably, the activity of an IncRNAs selected from SEQ ID NOs 23 to 42 is reduced by at least 50%, more preferred at least 75% such as at least 90% or 95%, even more preferred at least 98%, and most preferably about 100%. Means and methods for determining the reduction of activity of RNA are established in the art and are described, for example, in Esau et al. (2004), JBC, 279:52361-52365 or Gribbings et al. (2009), Nature Cell Biology 11 , 1143- 1149. Compounds as defined herein in item (i) may be an antisense molecule, siRNA, shRNA, antibody, ribozyme, aptamer, or small molecule. These and other compounds will be further detailed herein below. The efficiency of an inhibiting compound can be quantified by methods comparing the level of activity in the presence of the inhibitor to that in the absence of the inhibitor. For example, as an activity measure may be used: the change in amount of IncRNA formed. Such a method may be effected in high-throughput format in order to test the efficiency of several inhibiting compound simultaneously. High-throughput assays, independently of being biochemical, cellular or other assays, generally may be performed in wells of microtiter plates, wherein each plate may contain 96, 384 or 1536 wells. Handling of the plates, including incubation at temperatures other than ambient temperature, and bringing into contact of test compounds with the assay mixture is preferably effected by one or more computer-controlled robotic systems including pipetting devices. In case large libraries of test compounds are to be screened and/or screening is to be effected within short time, mixtures of, for example 10, 20, 30, 40, 50 or 100 test compounds may be added to each well. In case a well exhibits the expected activity, said mixture of test compounds may be de-convoluted to identify the one or more test compounds in said mixture giving rise to said activity.

As is evident from the examples herein below, IncRNAs which are involved in angiogenic signalling were identified in an experimental setup, wherein the expression of IncRNAs was determined in Human Umbilical Vein Endothelial cells (HUVEC cells) which have been cultured under hypoxic conditions (i.e. 0.2% oxygen for 24h) and compared to HUVEC control cells cultured under normal oxygen conditions. Hypoxia is a strong-pro-angiogenic stimulus (Pugh and Radcliffe (2003), Nat Med, 9(6):677-84). In simple terms, hypoxia drives tumor angiogenesis. The relationship between the two is often considered a matter of supply and demand. For example, ineffectively-vascularized tumor tissue becomes hypoxic, stimulating neoangiogenesis to improve the influx of oxygen, thereby diminishing the angiogenic drive (Moeller et al (2004), Semin Radiat Oncol; 14(3):215-21). Culturing HUVEC cells under hypoxic conditions is an established model for determining angiogenic responses and compounds involved in angiogenic signalling (for example, Veschini et al. (2007), Blood, 109: 2565-2570 and Calvani et al. (2007), Blood, 107(7):2705-2712).

The expression profile of IncRNAs under hypoxic conditions and normal oxygen (i.e. normoxic) conditions was determined by using the NCode Array technology as well as RNA- sequencing techniques (RNA-Seq). It was surprisingly found that certain IncRNAs are significantly differentially expressed after hypoxia in HUVECs. These IncRNAs are shown in Table 1. In more detail, IncRNAs corresponding to SEQ ID NO: 1 to 22 were found to be significantly upregulated under hypoxic conditions, while IncRNAs corresponding to SEQ ID NO: 23 to 42 were found to be significantly downregulated under hypoxic conditions. Because hypoxia is a strong-pro-angiogenic stimulus the IncRNAs of SEQ ID NOs 1 to 22 are pro- angiogenic factors. Hence, a compound promoting the expression and/or the activity of one or more long IncRNAs selected from SEQ ID NOs 1 to 22 will be beneficial in a pro-angiogenic therapy. On the other hand, IncRNAs of SEQ ID NO: 23 to 42 are anti-angiogenic factors. A compound inhibiting the expression and/or the activity of one or more (IncRNAs selected from SEQ ID NOs 23 to 42 will therefore be beneficial in a pro-angiogenic therapy. This principle has been further experimentally proven for the pro-angiogenic factors of SEQ ID NOs 1 to 3 and SEQ ID NO: 14.

SEQ ID NOs 1 to 3 are the three different isoforms of the IncRNA DSCAM-1. As defined herein, SEQ ID NO: 1 is the isoform 3, SEQ ID NO: 2 is the isoform 2, and SEQ ID NO: 3 is the isoform 1 of DSCAM-1. DSCAM-1 is also referred to herein as Iinc00323, Iinc323 or HSLINCR. The designation DSCAM-1 is in accordance with the LNCipedia database, the designation Iinc00323 or Iinc323 is in accordance with the Ensembl Genome Browser, and the designation HSLINCR is a designation of the inventors. SEQ ID NO: 1 (DSCAM-1 , isoform 3) is also referred to herein as linc00323-003, HSLINCR-003 or lnc-DSCAM-1 :2 (noting that in the LNCipedia database isoforms 2 and 3 are named the other way round as in the Ensembl Genome Browser). SEQ ID NO: 2 (DSCAM-1 , isoform 2) is also designated herein as linc00323-002, HSLINCR-002 or lnc-DSCAM-1 :3. SEQ ID NO: 1 (DSCAM-1 , isoform 1) is also named herein Iinc00323-001 , HSLINCR-001 or lnc-DSCAM-1 :1. All three isoforms share the sequence of SEQ ID NO: 43 and isoforms 2 and 3 additionally share the sequence of SEQ ID NO: 44 (see Figure 1 ). The IncRNA DSCAM-1 is encoded on chromosome 21. The expression of all three isoforms is significantly upregulated under hypoxic conditions, most significantly the expression of IncRNA DSCAM-1 isoform 3 (SEQ ID NO: 1) (see Figure 3). An siRNA directed against of IncRNA DSCAM-1 isoform 3 impairs the proliferation of ECs under normoxic conditions (see Figure 5). Furthermore, the overexpression of IncRNA DSCAM-1 isoform 3 in ECs cells under normoxic conditions influences the expression of known angiogenesis-related genes. The expression of known pro-angiogenic factors, such as ICAM- 1 and VEGF, was significantly increased upon the overexpression of DSCAM-1 isoform 3 in ECs under normoxic conditions (see Figure 9). In addition, VEGF stimulates the expression of DSCAM-1 isoform 3 (see Figure 10). This body of evidence confirms that the IncRNAs of SEQ ID NOs 1 to 3 are pro-angiogenic factors.

SEQ ID NO: 14 represents the IncRNA PLAC1-1. PLAC1-1 is also referred to in the LNCipedia database as NONHSAT138623 or NR_024607. Moreover, the IncRNA PLAC1-1 is named MIR503HG-002 in the Ensembl Genome Browser. The IncRNA PLAC1-1 is encoded on the X chromosome (see Figure 12A). The expression of the IncRNA PLAC1-1 is significantly upregulated under hypoxic conditions both in the cytoplasmic as well as the nuclear compartment (see Figure 11). When the IncRNA lnc-PLAC1-1 is silenced by a GapmeR construct in HUVECs scratch wound closure (and consequently angiogenesis) is impaired (see Figure 12E). Moreover, a GapmeR against the IncRNA lnc-PLAC1-1 represses GATA2 on protein level (see Figure 12G), noting that GATA2 is a key angiogenic factor. This body of evidence shows that also the IncRNA of SEQ ID NO: 14 is a pro-angiogenic factor.

Hypoxia is a strong pro-angiogenic stimulus and induces VEGF-mediated signalling in endothelial cells. The Examples provided herein below evidence that IncRNA expression in endothelial cells is strongly altered by hypoxia. The IncRNAs of SEQ ID NOs 1 to 42 were found to be highly sensitive towards hypoxia and to be crucial for endothelial angiogenic characteristics. As exemplarily shown for DSCAM-1 and PLAC1-1 silencing of these IncRNAs impairs function of endothelial cells via the block of proliferative pathways. To the best knowledge of the inventors none of the IncRNAs of SEQ ID NOs 1 to 42 has been described in endothelial biology and they have surprisingly been found to function as crucial factors to control endothelial cell behavior. The choice to further investigate DSCAM-1 and lnc-PLAC1-1 was dependent on their high deregulation in either microarray or RNA-Seq datasets. It can be expected that also the other so far not further characterized IncRNAs of SEQ ID NOs 4 to 13 and 15 to 42 are pro-angiogenic or anti-angiogenic factors which makes them likewise suitable for medical purposes, in particular the treatment of ischemia and the promotion of wound healing.

The present invention relates in a second aspect to a compound (i) promoting the expression and/or the activity of one or more IncRNAs selected from SEQ ID NOs 1 to 22, preferably selected from SEQ ID NOs 1 to 3 and 14; and/or (ii) inhibiting the expression and/or the activity of one or more IncRNAs selected from SEQ ID NOs 23 to 42 for use in treating or preventing ischemia, preferably cardiac ischemia and most preferably coronary artery disease, or for use in promoting wound healing. Compounds as defined herein in items (i) and (ii) have been detailed herein above in connection with the first aspect of the invention. The same compounds can be used in connection with the second aspect of the invention.

Ischemia is a restriction in blood supply to tissues, causing a shortage of oxygen and glucose needed for cellular metabolism (to keep tissue alive). Ischemia is a vascular disease involving an interruption in the arterial blood supply to a tissue, organ, or extremity that, if untreated, can lead to tissue death. It can be caused by embolism, thrombosis of an atherosclerosis artery, or trauma. Venous problems like venous outflow obstruction and low-flow states can cause acute arterial ischemia. An aneurysm is one of the most frequent causes of acute arterial ischemia. Other causes are heart conditions including myocardial infarction, mitral valve disease, chronic atrial fibrillation, cardiomyopathies, and prosthesis, in all of which thrombi are prone to develop. Hence, an ischemia can occur in several organs and tissues, such as brain, limbs, bowel or heart. Cardiac ischemia may be asymptomatic or may cause chest pain, known as angina pectoris. It occurs when the heart muscle, or myocardium, receives insufficient blood flow. This most frequently results from atherosclerosis, which is the long-term accumulation of cholesterol-rich plaques in the coronary arteries. Ischemic heart disease is the most common cause of death in most Western countries and a major cause of hospital admissions. Coronary artery disease (CAD) also known as atherosclerotic heart disease, coronary heart disease, or ischemic heart disease (IHD), is the most common type of heart disease and cause of heart attacks. The disease is caused by plaque building up along the inner walls of the arteries of the heart, which narrows the arteries and reduces blood flow to the heart. Pro-angiogenic therapy is a known means to treat or prevent ischemia (Marti and Risau (1999), Thromb Haemost. Sep;82 Suppl 1 :44-52).

Wound healing is the intricate process whereby the skin (or another organ-tissue) repairs itself after injury. The cell proliferation phase during wound healing is characterized by angiogenesis, collagen deposition, granulation tissue formation, epithelialization, and wound contraction (Midwood et al (2004), The International Journal of Biochemistry & Cell Biology; 36(6): 1031 -1037). In angiogenesis, vascular endothelial cells form new blood vessels. Hence, a pro-angiogenic therapy is a means to promote wound healing (Bahatia (2013), Mechanical and Chemical Signaling in Angiogenesis, Studies in Mechanobiology, Tissue Engineering and Biomaterials; 12(261-278)).

In accordance with a preferred embodiment of the first and second aspect of the invention the compound as defined in (i) is (a) a nucleic acid sequence which comprises or consists of the nucleic acid sequence of one or more IncRNAs selected from SEQ ID NOs 1 to 22 or an nucleic acid sequence which is at least 70% identical thereto, (b) an expression vector expressing the nucleic acid sequence as defined in (a), preferably under the control of a heart- specific promoter, or (c) a host comprising the expression vector of (b). As discussed above the three isoforms of SEQ ID NOs 1 to 3 share the sequence of SEQ ID NO: 43 and SEQ ID NO 1 and 2 additionally share the sequence of SEQ ID NO: 44. Hence, a nucleic acid sequence which is at least 70% identical to one of SEQ ID NOs 1 to 3 preferably retains the nucleotides corresponding to SEQ ID NO: 43 and more preferably a nucleic acid sequence which is at least 70% identical to one of SEQ ID NO 2 or 3 retains the nucleotides corresponding to SEQ ID NO: 43 and 44. In other terms, these nucleotides corresponding to SEQ ID NO: 43 or SEQ ID NO: 43 and 44 preferably remain unchanged and no amino acid changes are to be introduced in the subsequences.

The term "nucleic acid sequence" or "nucleotide sequence", in accordance with the present invention, includes DNA, such as cDNA or, in a preferred embodiment genomic DNA, and RNA. It is understood that the term "RNA" as used herein comprises all forms of RNA including, in a preferred embodiment, mRNA or miRNA. The term "nucleic acid sequence" is interchangeably used in accordance with the invention with the term "polynucleotide". The nucleic acid sequence according to item (a) of this preferred embodiment may be a recombinantly produced or isolated IncRNAs selected from SEQ ID NOs 1 to 22, any precursor thereof or any fragment thereof as long as a sequence identity of at least 70% over the entire length of an IncRNA selected from SEQ ID NOs 1 to 22 is maintained. Also orthologous or homologous sequences of the IncRNA selected from SEQ ID NOs 1 to 22 from different species including precursors or functional fragments thereof may be used. The fragments have to retain or essentially retain the function of the full-length IncRNA. Hence, the fragments have to be functional fragments.

The sequence identity of the nucleic acid sequence according to item (a) to an IncRNA selected from SEQ ID NOs 1 to 22 is with increasing preference at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99% and 100%. Means and methods for determining sequence identity are known in the art. Preferably, the BLAST (Basic Local Alignment Search Tool) program is used for determining the sequence identity with regard to one or more IncRNAs selected from SEQ ID NOs 1 to 22.

In accordance with items (b) and (c) of the above preferred embodiment such a compound may also be an expression vector or host being capable of producing an nucleic acid sequence as defined in item (a). An expression vector may be a plasmid that is used to introduce a specific transcript into a target cell. Once the expression vector is inside the cell, the protein that is encoded by the gene is produced by the cellular-transcription and translation machinery ribosomal complexes. The plasmid is in general engineered to contain regulatory sequences that act as enhancer and/or promoter regions and lead to efficient transcription of the transcript. In accordance with the present invention the expression vector preferably contains a heart-specific promoter. Heart-specific promoters are known in the art, for example, from Boecker at al. (2004), Mol W

12

Imagin.; 3(2):69-75. This ensures that the nucleic acid sequence is only expressed in the heart and may avoid potential unwanted side effects by expression in other organs.

Non-limiting examples of expression vectors include prokaryotic plasmid vectors, such as the pUC-series, pBluescript (Stratagene), the pET-series of expression vectors (Novagen) or pCRTOPO (Invitrogen) and vectors compatible with an expression in mammalian cells like pREP (Invitrogen), pcDNA3 (Invitrogen), pCEP4 (Invitrogen), pMCI neo (Stratagene), pXT1 (Stratagene), pSG5 (Stratagene), EBO-pSV2neo, pBPV-1 , pdBPVMMTneo, pRSVgpt, pRSVneo, pSV2-dhfr, plZD35, pLXIN, pSIR (Clontech), pIRES-EGFP (Clontech), pEAK-10 (Edge Biosystems) pTriEx-Hygro (Novagen) and pCINeo (Promega). Examples for plasmid vectors suitable for Pichia pastoris comprise e.g. the plasmids pA0815, pPIC9K and pPIC3.5K (all Intvitrogen). For the formulation of a pharmaceutical composition a suitable vector is selected in accordance with good manufacturing practice. Such vectors are known in the art, for example, from Ausubel et al, Hum Gene Ther. 2011 Apr; 22(4):489-97 or Allay et al., Hum Gene Ther. May 2011 ; 22(5): 595-604.

A typical mammalian expression vector contains the promoter element, which mediates the initiation of transcription of mRNA, the protein coding sequence, and signals required for the termination of transcription and polyadenylation of the transcript. Moreover, elements such as origin of replication, drug resistance gene, regulators (as part of an inducible promoter) may also be included. The lac promoter is a typical inducible promoter, useful for prokaryotic cells, which can be induced using the lactose analogue isopropylthiol-b-D-galactoside. ("IPTG"). For recombinant expression and secretion, the polynucleotide of interest may be ligated between e.g. the PelB leader signal, which directs the recombinant protein in the periplasm and the gene III in a phagemid called pHEN4 (described in Ghahroudi et al, 1997, FEBS Letters 414:521-526). Additional elements might include enhancers, Kozak sequences and intervening sequences flanked by donor and acceptor sites for RNA splicing. Highly efficient transcription can be achieved with the early and late promoters from SV40, the long terminal repeats (LTRs) from retroviruses, e.g., RSV, HTLVI, HIVI, and the early promoter of the cytomegalovirus (CMV). However, cellular elements can also be used (e.g., the human actin promoter). Suitable expression vectors for use in practicing the present invention include, for example, vectors such as pSVL and pMSG (Pharmacia, Uppsala, Sweden), pRSVcat (ATCC 37152), pSV2dhfr (ATCC 37146) and pBC12MI (ATCC 67109). Alternatively, the recombinant (poly)peptide can be expressed in stable cell lines that contain the gene construct integrated into a chromosome. The co-transfection with a selectable marker such as dhfr, gpt, neomycin, hygromycin allows the identification and isolation of the transfected cells. The transfected nucleic acid can also be amplified to express large amounts of the encoded (poly)peptide. The DHFR (dihydrofolate reductase) marker is useful to develop cell lines that carry several hundred or even several thousand copies of the gene of interest. Another useful selection marker is the enzyme glutamine synthase (GS) (Murphy et al.1991 , Biochem J. 227:277-279; Bebbington et al. 1992, Bio/Technology 10: 169-175). Using these markers, the mammalian cells are grown in selective medium and the cells with the highest resistance are selected. As indicated above, the expression vectors will preferably include at least one selectable marker. Such markers include dihydrofolate reductase, G418 or neomycin resistance for eukaryotic cell culture and tetracycline, kanamycin or ampicillin resistance genes for culturing in E. coli and other bacteria. For vector modification techniques, see Sambrook and Russel (2001 ), Molecular Cloning: A Laboratory Manual, 3 Vol.. Generally, vectors can contain one or more origins of replication (ori) and inheritance systems for cloning or expression, one or more markers for selection in the host, e.g., antibiotic resistance, and one or more expression cassettes. Suitable origins of replication (ori) include, for example, the Col E1 , the SV40 viral and the M 13 origins of replication.

The coding sequences inserted in the vector can e.g. be synthesized by standard methods, or isolated from natural sources. Ligation of the coding sequences to transcriptional regulatory elements and/or to other amino acid encoding sequences can be carried out using established methods. Transcriptional regulatory elements (parts of an expression cassette) ensuring expression in prokaryotes or eukaryotic cells are well known to those skilled in the art. These elements comprise regulatory sequences ensuring the initiation of the transcription (e.g., translation initiation codon, promoters, enhancers, and/or insulators), internal ribosomal entry sites (IRES) (Owens, Proc. Natl. Acad. Sci. USA 98 (2001), 1471-1476) and optionally poly-A signals ensuring termination of transcription and stabilization of the transcript. Additional regulatory elements may include transcriptional as well as translational enhancers, and/or naturally-associated or heterologous promoter regions. Preferably, the nucleotide sequence as defined in item (a) of the above preferred embodiment of the invention is operatively linked to such expression control sequences allowing expression in prokaryotic or eukaryotic cells.

The host may be a prokaryotic or eukaryotic cell. A suitable eukaryotic host may be a mammalian cell, an amphibian cell, a fish cell, an insect cell, a fungal cell or a plant cell. Representative examples of bacterial cells are E. coli, Streptomyces and Salmonella typhimurium cells; of fungal cells are yeast cells; and of insect cells are Drosophila S2 and Spodoptera Sf9 cells. It is preferred that the cell is a mammalian cell such as a human cell. Mammalian host cells that could be used include, human Hela, 293, H9 and Jurkat cells, mouse NIH3T3 and C127 cells, Cos 1 , Cos 7 and CV1 , quail QC1-3 cells, mouse L cells and Chinese hamster ovary (CHO) cells. The cell may be a part of a cell line, preferably a human cell line. Appropriate culture mediums and conditions for the above-described host cells are known in the art. The host is preferably a host cell and more preferably an isolated host cell. The host is also preferably a non-human host.

In accordance with another preferred embodiment of the first and second aspect of the invention the compound as defined in (i) is (a) a transcription factor promoting the expression of one or more IncRNAs selected from SEQ ID NOs 1 to 22, and/or (b) a small molecule enhancing the expression of one or more IncRNAs selected from SEQ ID NOs 1 to 22.

The term "transcription factor" as used is connection with this embodiment defines a protein or peptide that binds to specific DNA sequences, thereby controlling the transcription of the genes encoding of one or more IncRNAs selected from SEQ ID NOs 1 to 22. The efficiency of a transcription factor in activating the expression of an IncRNA selected from SEQ ID NOs 1 to 22 can be quantified by methods comparing the level of the IncRNA in the presence of the transcription factor to that in the absence of the transcription factor. For example, as an activity measure the change in amount of IncRNA formed may be used. Such a method may be effected in high-throughput format in order to test the efficiency of several inhibiting compound simultaneously. High-throughput formats have been further detailed herein above.

The small molecule enhancing the expression of one or more IncRNAs selected from SEQ ID NOs 1 to 22 is a low molecular weight organic compound which is by definition not a polymer. The small molecule of the invention is preferably a molecule that binds with high affinity to an IncRNA of SEQ ID NOs 1 to 22 and in addition enhances the activity of an IncRNA of SEQ ID NOs 1 to 22. The upper molecular weight limit for a small molecule is preferably 1500Da, more preferably 10OODa and most preferably 800Da which allows for the possibility to rapidly diffuse across cell membranes so that they can reach intracellular sites of action. Libraries of small organic molecules and high-throughput techniques for screening such libraries with a specific target molecule, in the present case an IncRNA selected from SEQ ID NOs 1 to 22, are established in the art.

In accordance with a further preferred embodiment of the first and second aspect of the invention the compound as defined in (ii) is (a) a nucleic acid sequence which comprises or consists of a nucleotide sequence being complementary to at least 12 continuous nucleotides of a IncRNAs selected from SEQ ID NOs 23 to 42, (b) a nucleic acid sequence which comprises or consists of a nucleotide sequence which is at least 70% identical to the complementary strand of one or more IncRNAs selected from SEQ ID NOs 23 to 42, (c) a nucleic acid sequence which comprises or consists of a nucleotide sequence according to (a) or (b), wherein U is replaced by T, (d) an expression vector expressing the nucleic acid sequence as defined in any one of (a) to (c), preferably under the control of a heart-specific promoter, or (e)a host comprising the expression vector of (d).

The nucleic acid sequences as defined in items (a) to (c) of this preferred embodiment comprise or consist of sequences that comprise or are complementary to nucleotides of a IncRNAs selected from SEQ ID NOs 23 to 42. Hence, these nucleic acid sequences comprise or are antisense nucleic acid sequences. The antisense technology for silencing the expression of a target gene is well-established and widely used in the art to treat various diseases. The molecule according to item (a) of this preferred embodiment of the invention comprises or consists of a sequence which is with increasing preference complementary to at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, or all 23 nucleotides of SEQ ID NOs 23 to 42. These at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, or at least 21 nucleotides are preferably a contiguous part of SEQ ID NOs 23 to 42 i.e. the nucleotides are consecutive in the respective SEQ ID NO. The molecule according to item (a) is preferably a "siRNA". The term "siRNA" in accordance with the present invention refers to small interfering RNA, also known as short interfering RNA or silencing RNA. siRNAs are a class of 18 to 30, preferably 20 to 25, most preferred 21 to 23 or 21 nucleotide-long double-stranded RNA molecules that play a variety of roles in biology. Most notably, siRNA is involved in the RNA interference (RNAi) pathway where the siRNA interferes with the expression of a specific gene. In addition to their role in the RNAi pathway, siRNAs also act in RNAi-related pathways, e.g. as an antiviral mechanism or in shaping the chromatin structure of a genome. siRNAs have a well defined structure: a short double-strand of RNA (dsRNA), advantageously with at least one RNA strand having an overhang. Each strand typically has a 5' phosphate group and a 3' hydroxyl (-OH) group. This structure is the result of processing by dicer, an enzyme that converts either long dsRNAs or small hairpin RNAs into siRNAs. siRNAs can also be exogenously (artificially) introduced into cells to bring W

16 about the specific knockdown of a gene of interest. Thus, any gene of which the sequence is known can in principle be targeted based on sequence complementarity with an appropriately tailored siRNA. The double-stranded RNA molecule or a metabolic processing product thereof is capable of mediating target-specific nucleic acid modifications, particularly RNA interference and/or DNA methylation. Also preferably at least one RNA strand has a 5'- and/or 3' -overhang. Preferably, one or both ends of the double-strand has a 3'-overhang from 1-5 nucleotides, more preferably from 1-3 nucleotides and most preferably 2 nucleotides. In general, any RNA molecule suitable to act as siRNA is envisioned in the present invention. The most efficient silencing was so far obtained with siRNA duplexes composed of 21 -nt sense and 21 -nt antisense strands, paired in a manner to have 2-nt 3'- overhangs. The sequence of the 2-nt 3' overhang makes a small contribution to the specificity of target recognition restricted to the unpaired nucleotide adjacent to the first base pair (Elbashir et al. Nature. 2001 May 24;411(6836):494-8). 2'-deoxynucleotides in the 3' overhangs are as efficient as ribonucleotides, but are often cheaper to synthesize and probably more nuclease resistant. The siRNA according to the invention comprises an antisense strand which comprises or consists of a sequence which is with increasing preference complementary to at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least

17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, or all 23 nucleotides of SEQ ID NOs 23 to 42. These at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, or at least 21 nucleotides are preferably a contiguous part of SEQ ID NOs 23 to 42, i.e. the nucleotides are consecutive in the respective SEQ ID NO. The molecule according to item (a) is also preferably a "shRNA". A "shRNA" in accordance with the present invention is a short hairpin RNA, which is a sequence of RNA that makes a (tight) hairpin turn that can also be used to silence gene expression via RNA interference. shRNA preferably utilizes the U6 promoter for its expression. The shRNA hairpin structure is cleaved by the cellular machinery into siRNA, which is then bound to the RNA-induced silencing complex (RISC). This complex binds to and cleaves mRNAs which match the shRNA that is bound to it. The shRNA according to the invention comprises or consists a sequence which is with increasing preference complementary to at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least

18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, or all 23 nucleotides of SEQ ID NOs 23 to 42. These at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, or at least 21 nucleotides are preferably a contiguous part of SEQ ID NOs 23 to 42, i.e. the nucleotides are consecutive in the respective SEQ ID NO. An molecule according to item (b) of the above preferred embodiment of the invention is capable of interacting with, more specifically hybridizing with the target IncRNA. By formation of the hybrid the function of the IncRNA is reduced or blocked. Standard methods relating to such antisense technology have been described (see, e.g., Melani et al., Cancer Res. (1991 ) 51 :2897-2901 ). The term "antisense molecule" in accordance with the present invention thus relates to a nucleic acid molecule, preferably a RNA molecule, that has a base sequence complementary to a given IncRNA, i.e. the "sense" sequence.

A particularly preferred example of the molecule according to item (b) is an Endoribonuclease- prepared siRNA (esiRNA). An esiRNA is a mixture of siRNA oligos resulting from cleavage of a long double-stranded RNA (dsRNA) according to item (b) with an endoribonuclease such as Escherichia coli RNase III or dicer. esiRNAs are an alternative concept to the usage of chemically synthesized siRNA for RNA Interference (RNAi). An esiRNAs is the enzymatic digestion of a long double stranded RNA in vitro. For the generation of esiRNAs a cDNA of an IncRNA template may be amplified by PCR and tagged with two bacteriophage-promotor sequences. RNA polymerase is then used to generate long double stranded RNA that is complentary to the target-gene cDNA. This complentary RNA may be subsequently digested with RNase III from Escherichia coli to generate short overlapping fragments of siRNAs with a length between 18-25 base pairs. This complex mixture of short double stranded RNAs is similar to the mixture generated by Dicer cleavage in vivo and is therefore called endoribonuclease-prepared siRNA or short esiRNA. Hence, esiRNA are a heterogeneous mixture of siRNAs that all target the same mRNA sequence. esiRNAs lead to highly specific and effective gene silencing.

The sequence identity of the antisense molecule according to item (b) to an IncRNA selected from SEQ ID NOs 23 to 42 is with increasing preference at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99% and 100%. Means and methods for determining sequence identity are known in the art. Preferably, the BLAST (Basic Local Alignment Search Tool) program is used for determining the sequence identity with regard to one or more IncRNAs selected from SEQ ID NOs 23 to 42. Antisense molecules, siRNAs and shRNAs of the present invention are preferably chemically synthesized using appropriately protected ribonucleoside phosphoramidites and a conventional RNA synthesizer. Suppliers of RNA synthesis reagents include Proligo (Hamburg, Germany), Dharmacon Research (Lafayette, CO, USA), Pierce Chemical (part of Perbio Science, Rockford, IL , USA), Glen Research (Sterling, VA, USA), ChemGenes (Ashland, MA, USA), and Cruachem (Glasgow, UK).

The ability of antisense molecules, siRNA, and shRNA to potently, but reversibly, silence IncRNA and genes in vivo makes these molecules particularly well suited for use in the pharmaceutical composition of the invention. Ways of administering siRNA to humans are described in De Fougerolles et al., Current Opinion in Pharmacology, 2008, 8:280-285. Such ways are also suitable for administering other small RNA molecules like shRNA. Accordingly, such pharmaceutical compositions may be administered directly formulated as a saline, via liposome based and polymer-based nanoparticle approaches, as conjugated or complexation pharmaceutical compositions, or via viral delivery systems. Direct administration comprises injection into tissue, intranasal and intratracheal administration. Liposome based and polymer- based nanoparticle approaches comprise the cationic lipid Genzyme Lipid (GL) 67, cationic liposomes, chitosan nanoparticles and cationic cell penetrating peptides (CPPs). Conjugated or complexation pharmaceutical compositions comprise PEI-complexed antisense molecules, siRNA, shRNA or miRNA. Further, viral delivery systems comprise influenza virus envelopes and virosomes.

The antisense molecules, siRNAs, shRNAs may comprise modified nucleotides such as locked nucleic acids (LNAs). The ribose moiety of an LNA nucleotide is modified with an extra bridge connecting the 2' oxygen and 4' carbon. The bridge "locks" the ribose in the 3'-endo (North) conformation, which is often found in the A-form duplexes. LNA nucleotides can be mixed with DNA or RNA residues in the oligonucleotide whenever desired. Such oligomers are synthesized chemically and are commercially available. The locked ribose conformation enhances base stacking and backbone pre-organization. This significantly increases the hybridization properties (melting temperature) of oligonucleotides. Particularly preferred example of siRNAs is GapmeR (LNA™ GapmeRs (Exiqon)). GapmeRs are potent antisense oligonucleotides used for highly efficient inhibition of mRNA and IncRNA function. GapmeRs contain a central stretch of DNA monomers flanked by blocks of LNAs. The GapmeRs are preferably 14-16 nucleotides in length and are optionally fully phosphorothioated. The DNA gap activates the RNAse H-mediated degradation of targeted RNAs and is also suitable to target transcripts directly in the nucleus. GapmeRs are used in the examples, e.g., to downregulate the IncRNA DSCAM-1 isoform 3 (LINC00323-003) (SEQ ID NO: 1 ) in HUVEC cells.

Examples of suitable expression vectors which may be used in connection with item (d) of the above-preferred embodiment have been detailed herein above.

In accordance with a different preferred embodiment of the first and second aspect of the invention the compound as defined in (ii) is an aptamer, a ribozyme, an antibody, a protein drug, or a small molecule inhibitor.

The aptamer, ribozyme, antibody, protein drug, or small molecule inhibitor of this embodiment specifically bind to one or more IncRNA selected from SEQ ID NOs 23 to 42, thereby inhibiting the activity of one or more IncRNA selected from SEQ ID NOs 23 to 42. The term "aptamer" in accordance with the present invention refers to DNA or RNA molecules being either in the natural D-conformation or in the L-conformation ("spiegelmer") that have been selected from random pools based on their ability to bind other molecules. Aptamers have been selected which bind nucleic acid, proteins, small organic compounds, and even entire organisms. A database of aptamers is maintained at http://aptamer.icmb.utexas.edu/. More specifically, aptamers can be classified as DNA or RNA aptamers or peptide aptamers. Whereas the former consist of (usually short) strands of oligonucleotides, the latter consist of a short variable peptide domain, attached at both ends to a protein scaffold. Nucleic acid aptamers are nucleic acid species that have been engineered through repeated rounds of in vitro selection or equivalently, SELEX (systematic evolution of ligands by exponential enrichment) to bind to various molecular targets such as small molecules, proteins, nucleic acids, and even cells, tissues and organisms. The molecular target envisaged by the present invention is a nucleic acid, namely an IncRNA selected from 23 to 42. Hence, aptamers can be produced against the target molecule of the invention. Peptide aptamers are peptides that are designed to interfere with other protein interactions inside cells. They consist of a variable peptide loop attached at both ends to a protein scaffold. This double structural constraint greatly increases the binding affinity of the peptide aptamer to levels comparable to an antibody's (nanomolar range). The variable loop length is typically comprised of 10 to 20 amino acids, and the scaffold may be any protein which has good solubility properties. Currently, the bacterial protein Thioredoxin-A is the most used scaffold protein, the variable loop being inserted within the reducing active site, which is a -Cys-Gly-Pro-Cys- loop in the wild protein, the two cysteins lateral chains being able to form a disulfide bridge. Peptide aptamer selection can be made using different systems, but the most used is currently the yeast two-hybrid system.

Aptamers offer the utility for biotechnological and therapeutic applications as they offer molecular recognition properties that rival those of the commonly used biomolecules, in particular antibodies. In addition to their discriminate recognition, aptamers offer advantages over antibodies as they can be engineered completely in a test tube, are readily produced by chemical synthesis, possess desirable storage properties, and elicit little or no immunogenicity in therapeutic applications. Non-modified aptamers are cleared rapidly from the bloodstream, with a half-life of minutes to hours, mainly due to nuclease degradation and clearance from the body by the kidneys, a result of the aptamer's inherently low molecular weight. The rapid clearance of aptamers can be an advantage in applications such as in vivo diagnostic imaging. Several modifications, such as 2'-fluorine-substituted pyrimidines, polyethylene glycol (PEG) linkage, etc. are available to scientists with which the half-life of aptamers easily can be increased to the day or even week time scale.

The term "ribozymes" refers to RNA molecules that act as enzymes in the absence of proteins. These RNA molecules act catalytic or autocatalytic and are capable of cleaving e.g. other RNAs at specific target sites but they have also been found to catalyze the aminotransferase activity of the ribosome. Selection of appropriate target sites and corresponding ribozymes can be done as described for example in Zaher and Unrau (2007), RNA 13 (7): 1017-1026.

Examples of well-characterized small self-cleaving RNAs are the hammerhead, hairpin, hepatitis delta virus, and in vitro-selected lead-dependent ribozymes. The organization of these small catalysts is in contrast to that of larger ribozymes, such as the group I intron.

The principle of catalytic self-cleavage has become well established in the last 10 years. The hammerhead ribozymes are characterized best among the RNA molecules with ribozyme activity. Since it was shown that hammerhead structures can be integrated into heterologous RNA sequences and that ribozyme activity can thereby be transferred to these molecules, it appears that catalytic sequences for almost any target sequence can be created, provided the target sequence contains a potential matching cleavage site.

The basic principle of constructing hammerhead ribozymes is as follows: An interesting region of the RNA, which contains the GUC (or CUC) triplet, is selected. Two oligonucleotide strands, each with 6 to 8 nucleotides, are taken and the catalytic hammerhead sequence is inserted between them. Molecules of this type were synthesized for numerous target sequences. They showed catalytic activity in vitro and in some cases also in vivo. The best results are usually obtained with short ribozymes and target sequences. Since the target sequence is a short RNA sequence, namely an IncRNA selected from SEQ ID NOs 23 to 42. IncRNAs selected from SEQ ID NOs 23 to 42 are bona fide targets sequences for the generation of ribozymes being capable to specifically cleave an IncRNA selected from SEQ ID NOs 23 to 42. Also the aptamers and ribozymes may comprise modified nucleotides, such as locked nucleic acids (LNAs).

The term "antibody" as used in accordance with the present invention comprises, for example, polyclonal or monoclonal antibodies. Furthermore, also derivatives or fragments thereof, which still retain the binding specificity, are comprised in the term "antibody". Antibody fragments or derivatives comprise, inter alia, Fab or Fab' fragments, Fd, F(ab') 2 , Fv or scFv fragments, single domain V H or V-like domains, such as VhH or V-NAR-domains, as well as multimeric formats such as minibodies, diabodies, tribodies, tetrabodies or chemically conjugated Fab'- multimers (see, for example, Altshuler et al., 2010., Holliger and Hudson, 2005). The term "antibody" also includes embodiments such as chimeric (human constant domain, non-human variable domain), single chain and humanized (human antibody with the exception of non- human CDRs) antibodies.

Various techniques for the production of antibodies and fragments thereof are well known in the art and described, e.g. in Altshuler et al., 2010. Thus, polyclonal antibodies can be obtained from the blood of an animal following immunisation with an antigen in mixture with additives and adjuvans and monoclonal antibodies can be produced by any technique which provides antibodies produced by continuous cell line cultures. Examples for such techniques are described, e.g. Harlow and Lane (1988) and (1999) and include the hybridoma technique originally described by Kohler and Milstein, 1975, the trioma technique, the human B-cell hybridoma technique (see e.g. Kozbor, 1983; Li et al., 2006) and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole et al., 1985). Furthermore, recombinant antibodies may be obtained from monoclonal antibodies or can be prepared de novo using various display methods such as phage, ribosomal, mRNA, or cell display. A suitable system for the expression of the recombinant (humanized) antibodies or fragments thereof may be selected from, for example, bacteria, yeast, insects, mammalian cell lines or transgenic animals or plants (see, e.g., US patent 6,080,560; Holliger and Hudson, 2005). Further, techniques described for the production of single chain antibodies (see, inter alia, US Patent 4,946,778) can be adapted to produce single chain antibodies specific for the target of this invention. Surface plasmon resonance as employed in the BIAcore system can be used to increase the efficiency of phage antibodies. The term "protein drugs" designates designer drugs that are derivatives of human proteins. These proteins are used as scaffold to create a protein drug by well-established screening procedures (see Tomlinson et al (2004), NATURE BIOTECHNOLOGY, 22(5): 521-522). Non- limiting examples of human proteins which serve as a scaffold for designing protein drugs are transferrin, C-type lectins, trinectins, domain antibodies, kunitz domains, lipocalins and the Fyn SH3 domain.

A small molecule inhibitor is a low molecular weight organic compound which is by definition not a polymer. The small molecule of the invention is preferably a molecule that binds with high affinity to an IncRNA of SEQ ID NOs 2 to 42 and in addition inhibits the activity of an IncRNA of SEQ ID NOs 2 to 42. The upper molecular weight limit for a small molecule is preferably 1500Da, more preferably 10OODa and most preferably 800Da which allows for the possibility to rapidly diffuse across cell membranes so that they can reach intracellular sites of action. Libraries of small organic molecules and high-throughput techniques for screening such libraries with a specific target molecule, in the present case an IncRNA selected from SEQ ID NOs 2 to 42, are established in the art.

Antisense molecule, siRNA, shRNA, antibody, enzyme, ribozyme, aptamer, protein drug, or small molecule inhibitor may be fused to a lipid, such as a cholesterol. Means and methods to introduce lipid modifications and in particular a cholesterol modification to a nucleic acid molecule are described in Kriitzfeldt et al. 2005 (Nature 438, 685-689). For example, a cholesterol may be linked through a hydroxylprolinol linkage to a nucleic acid molecule. Such modifications increase the efficiency of the uptake of a nucleic acid molecule and in particular of small RNAs into the cell.

The present invention relates in a third aspect to a pharmaceutical composition comprising (i) a compound promoting the expression and/or the activity of one or more IncRNAs selected from SEQ ID NOs 23 to 42; and/or (ii) a compound inhibiting the expression and/or the activity of one or more long non-coding RNAs (IncRNAs) selected from SEQ ID NOs 1 to 22, preferably selected from SEQ ID NOs 1 to 3 and 14.

The formulation and administration of the pharmaceutical composition of the invention have been detailed herein above in connection with the first aspect of the invention. The same formulations and administrations can be used in connection with the second aspect of the invention. The composition of the third aspect of the invention is an anti-angiogenic pharmaceutical composition.

A compound promoting the expression of one or more IncRNAs selected from SEQ ID NOs 23 to 42 - as defined herein in item (i) - may be any compound enhancing or upregulating the transcription of an IncRNA selected from SEQ ID NOs 23 to 42. Non-limiting examples of such compounds are transcription factors enhancing the transcription of the genes encoding the IncRNAs selected from SEQ ID NOs 23 to 42 or a small molecule enhancing the expression of one or more IncRNAs selected from SEQ ID NOs 23 to 42. A transcription factor is a protein binding to specific DNA sequences, thereby controlling the transcription of genetic information from DNA to RNA. A small molecule is a low molecular weight compound which is by definition not a polymer. A compound promoting the activity of one or more IncRNAs selected from SEQ ID NOs 23 to 42 - as defined herein in item (i) - may be any compound which causes that said IncRNA effectively performs its function in a cell. Hence, in the simplest form such a compound may be a recombinantly produced or isolated IncRNA selected from SEQ ID NOs 23 to 42 or any precursor or fragment thereof. In this embodiment the administration of an recombinantly produced or isolated IncRNA increases the concentration of IncRNA in the subject to be treated. This higher concentration promotes the overall activity of the respective IncRNA in the subject. The fragments have to retain or essentially retain the function of the full-length IncRNA. Such a compound may also be a vector or host being capable of producing such an IncRNA. Hence, the fragments have to be functional fragments. Also orthologous or homologous sequences of the IncRNAs selected from SEQ ID NOs 23 to 42 from different species including precursors or functional fragments thereof may be used. Alternatively, such a compound may be a compound maintaining or even enhancing the activity of an IncRNA selected from SEQ ID NOs 23 to 42 by either directly or indirectly interacting with the IncRNA. For instance, such a compound may prevent an IncRNA selected from SEQ ID NOs 23 to 42 from degeneration by RNases or may be an interaction partner, such as another IncRNA, which binds to and promotes the activity of an IncRNA selected from SEQ ID NOs 23 to 42. Compounds as defined herein in item (i) will be further detailed herein below.

The efficiency of a compound as defined herein in item (ii) can also be quantified by methods comparing the level of expression and/or activity of an IncRNA selected from SEQ ID NOs 23 to 42 in the presence of an promotor of the IncRNA, such as a transcription factor, to that in the absence of the promotor. For example, as an activity measure the change in amount of IncRNA formed may be used. The method is preferably effected in high-throughput format as further detailed herein below.

A compound inhibiting the expression of one or more IncRNAs selected from SEQ ID NOs 1 to 22 - as defined herein in item (ii) - is in accordance with the present invention a compound lowering or preventing the transcription of one or more of the genes encoding the IncRNAs selected of SEQ ID NOs 1 to 22. Such compounds include compounds interfering with the transcriptional machinery and/or its interaction with the promoter of said genes and/or with expression control elements remote from the promoter such as enhancers. The compound inhibiting the expression of an IncRNAs selected from SEQ ID NOs 1 to 22 specifically inhibits the expression of said IncRNA, for example, by specifically interfering with the promoter region controlling the expression of the IncRNA. Preferably, the transcription of an IncRNAs selected from SEQ ID NOs 1 to 22 is reduced by at least 50%, more preferred at least 75% such as at least 90% or 95%, even more preferred at least 98% and most preferred by about 100%. A compound inhibiting the activity of an IncRNA selected from SEQ ID NOs 1 to 22 - as defined herein in item (ii) - in accordance with the present invention causes said IncRNA to perform its function with lowered efficiency. The compound inhibiting the activity of an IncRNA selected from SEQ ID NOs 1 to 22 specifically inhibits the activity of said IncRNA. Preferably, the activity of an IncRNA selected from SEQ ID NOs 1 to 22 is reduced by at least 50%, more preferred at least 75% such as at least 90% or 95%, even more preferred at least 98%, and most preferably about 100%. Means and methods for determining the reduction of activity of an RNA are established in the art and are described, for example, in Esau et al. (2004), JBC, 279:52361-52365 or Gribbings et al. (2009), Nature Cell Biology 11 , 1143-1149. Compounds as defined herein in item (i) may be an antisense molecule, siRNA, shRNA, antibody, ribozyme, aptamer, or small molecule. These and other compounds will be further detailed herein below.

As discussed above, the three isoforms of SEQ ID NOs 1 to 3 share the sequence of SEQ ID NO: 43 and SEQ ID NO 1 and 2 additionally share the sequence of SEQ ID NO: 44. Hence, a compound specifically inhibiting the activity of the SEQ ID NOs 1 to 3 may be designed by targeting SEQ ID NO: 43, and a compound specifically inhibiting the activity of the SEQ ID NOs 1 and 2 may be designed by targeting SEQ ID NO: 44.

The efficiency of an inhibiting compound can be quantified by methods comparing the level of activity in the presence of the inhibitor to that in the absence of the inhibitor. For example, as an activity measure the change in amount of IncRNA formed may be used. Such a method may be effected in high-throughput format in order to test the efficiency of several inhibiting compound simultaneously. High-throughput assays, independently of being biochemical, cellular or other assays, generally may be performed in wells of microtiter plates, wherein each plate may contain 96, 384 or 1536 wells. Handling of the plates, including incubation at temperatures other than ambient temperature, and bringing into contact of test compounds with the assay mixture is preferably effected by one or more computer-controlled robotic systems including pipetting devices. In case large libraries of test compounds are to be screened and/or screening is to be effected within short time, mixtures of, for example 10, 20, 30, 40, 50 or 100 test compounds may be added to each well. In case a well exhibits the expected activity, said mixture of test compounds may be de-convoluted to identify the one or more test compounds in said mixture giving rise to said activity.

As discussed in detail in connection with the first aspect of the invention, the IncRNAs of SEQ ID NOs 1 to 22 are pro-angiogenic factors. Hence, a compound inhibiting the expression and/or the activity of one or more IncRNAs selected from SEQ ID NOs 1 to 22 will be beneficial in an anti-angiogenic therapy. IncRNAs of SEQ ID NO: 23 to 42 are anti-angiogenic factors. Consequently, a compound promoting the expression and/or the activity of one or more IncRNAs selected from SEQ ID NOs 23 to 42 will be beneficial in an anti-angiogenic therapy. The present invention relates in a fourth aspect to a compound (i) promoting the expression and/or the activity of one or more IncRNAs selected from SEQ ID NOs 23 to 42; and/or (ii) inhibiting the expression and/or the activity of one or more IncRNAs selected from SEQ ID NOs 1 to 22, preferably selected from SEQ ID NOs 1 to 3 and 14 for use in treating or preventing a tumor, preferably a hypoxic tumor, or for use in treating or preventing diabetic retinopathy.

Compounds as defined herein in items (i) and (ii) have been detailed herein above in connection with the third aspect of the invention. The same compounds can be used in connection with the fourth aspect of the invention.

The terms "tumor" and "cancer" are interchangeably used herein. Angiogenesis plays a critical role in the growth and spread of cancer (Nishida et al. (2006), Vase Health Risk Management, 2(3): 213-219). A blood supply is necessary for tumors to grow beyond a few millimeters in size. Tumors can cause this blood supply to form by giving off chemical signals that stimulate angiogenesis. Tumors can also stimulate nearby normal cells to produce angiogenesis signalling molecules. The resulting new blood vessels "feed" growing tumors with oxygen and nutrients, allowing the cancer cells to invade nearby tissue, to move throughout the body, and to form new colonies of cancer cells, called metastases. Because tumors cannot grow beyond a certain size or spread without a blood supply, blocking tumor angiogenesis is a known anticancer therapy. Diabetic retinopathy is retinopathy (damage to the retina) caused by complications of diabetes, which can eventually lead to blindness. As the disease progresses, severe nonproliferative diabetic retinopathy enters an advanced, or proliferative (PDR) stage. The blood vessels proliferate in order to supplement the lack of oxygen in the retina caused by the cell proliferation. The growth of the blood vessels may be inhibited by an anti-angiogenic therapy (Bahatia (2013), Mechanical and Chemical Signaling in Angiogenesis, Studies in Mechanobiology, Tissue Engineering and Biomaterials; 12(261-278)).

In accordance with a preferred embodiment of the third and fourth aspect of the invention the compound as defined in (i) is (a) a nucleic acid sequence which comprises or consists of the nucleic acid sequence of one or more IncRNAs selected from SEQ ID NOs 23 to 42 or an nucleic acid sequence which is at least 70% identical thereto, (b) an expression vector expressing the nucleic acid sequence as defined in (a), preferably under the control of a heart- specific promoter, or (c) a host comprising the expression vector of (b). The nucleic acid sequence according to item (a) of this preferred embodiment may be a recombinantly produced or isolated IncRNA selected from SEQ ID NOs 23 to 42, any precursor thereof or any fragment thereof as long as a sequence identity of at least 70% over the entire length of an IncRNA selected from SEQ ID NOs 23 to 42 is maintained. Also orthologous or homologous sequences of the IncRNAs selected from SEQ ID NOs 23 to 42 from different species including precursors or a functional fragment thereof may be used. The fragments have to retain or essentially retain the function of the full-length IncRNA. Hence, the fragments have to be functional fragments.

The sequence identity of the nucleic acid sequence according to item (a) to an IncRNA selected from SEQ ID NOs 23 to 42 is with increasing preference at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99% and 100%. Means and methods for determining sequence identity are known in the art. Preferably, the BLAST (Basic Local Alignment Search Tool) program is used for determining the sequence identity with regard to one or more IncRNAs selected from SEQ ID NOs 23 to 42. In accordance with items (b) and (c) of the above preferred embodiment such a compound may also be an expression vector or host being capable of producing an nucleic acid sequence as defined in item (a). Suitable expression vectors and hosts have been detailed herein above in connection with the first and second aspect of the invention. These expression vectors can also be used in connection with the third and fourth aspect of the invention.

In accordance with another preferred embodiment of the third and fourth aspect of the invention the compound as defined in (i) is (a) a transcription factor promoting the expression of one or more IncRNAs selected from SEQ ID NOs 23 to 42, and/or (b) a small molecule enhancing the expression of one or more IncRNAs selected from SEQ ID NOs 23 to 42.

The term "transcription factor" as used is connection with this embodiment defines a protein or peptide that binds to specific DNA sequences, thereby controlling the transcription of the genes encoding of one or more IncRNAs selected from SEQ ID NOs 23 to 42. The efficiency of a transcription factor in activating the expression of an IncRNA selected from SEQ ID NOs 23 to 42 can be quantified by methods comparing the level of the IncRNA in the presence of the transcription factor to that in the absence of the transcription factor. For example, as an activity measure the change in amount of IncRNA formed may be used. Such a method may be effected in high-throughput format in order to test the efficiency of several inhibiting compound simultaneously. High-throughput formats have been further detailed herein above.

The small molecule enhancing the expression of one or more IncRNAs selected from SEQ ID NOs 23 to 42 is a low molecular weight organic compound which is by definition not a polymer. The small molecule of the invention is preferably a molecule that binds with high affinity to an IncRNA of SEQ ID NOs 23 to 42 and in addition enhances the activity of an IncRNA of SEQ ID NOs 23 to 42. The upper molecular weight limit for a small molecule is preferably 1500Da, more preferably 1000Da and most preferably 800Da which allows for the possibility to rapidly diffuse across cell membranes so that they can reach intracellular sites of action. Libraries of small organic molecules and high-throughput techniques for screening such libraries with a specific target molecule, in the present case an IncRNA selected from SEQ ID NOs 23 to 42, are established in the art.

In accordance with a preferred embodiment of the third and fourth aspect of the invention the compound as defined in (ii) is (a) a nucleic acid sequence which comprises or consists of a nucleotide sequence being complementary to at least 12 continuous nucleotides of a IncRNAs selected from SEQ ID NOs 1 to 22, (b) a nucleic acid sequence which comprises or consists of a nucleotide sequence which is at least 70% identical to the complementary strand of one or more IncRNAs selected from SEQ ID NOs 1 to 22, (c) a nucleic acid sequence which comprises or consists of a nucleotide sequence according to (a) or (b), wherein U is replaced by T, (d) an expression vector expressing the nucleic acid sequence as defined in any one of (a) to (c), preferably under the control of a heart-specific promoter, or (e) a host comprising the expression vector of (d).

The nucleic acid sequences as defined in items (a) to (c) of this preferred embodiment comprise or consist of sequences that comprise or are complementary to nucleotides of a IncRNAs selected from SEQ ID NOs 1 to 22. Hence, these nucleic acid sequences comprise or are antisense nucleic acid sequences.

The molecule according to item (a) of this preferred embodiment of the invention comprises or consists of a sequence which is with increasing preference complementary to at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least

17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, or all 23 nucleotides of SEQ ID NOs 1 to 22.

These at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least

20 nucleotides, or at least 21 nucleotides are preferably a contiguous part of SEQ ID NOs 1 to

22, i.e. the nucleotides are consecutive in the respective SEQ ID NO.

The molecule according to item (a) is preferably a "siRNA". siRNAs are a class of 18 to 30, preferably 20 to 25, most preferred 21 to 23 or 21 nucleotide-long double-stranded RNA molecules that play a variety of roles in biology. Also preferably at least one RNA strand has a 5'- and/or 3'-overhang. Preferably, one or both ends of the double-strand have a 3'-overhang from 1-5 nucleotides, more preferably from 1-3 nucleotides and most preferably 2 nucleotides. In general, any RNA molecule suitable to act as siRNA is envisioned in the present invention. The most efficient silencing was so far obtained with siRNA duplexes composed of 21-nt sense and 21-nt antisense strands, paired in a manner to have 2-nt 3'- overhangs. The siRNA according to the invention comprises an antisense strand which comprises or consists of a sequence which is with increasing preference complementary to at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, or all 23 nucleotides of SEQ ID NOs 1 to 22. These at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, or at least 21 nucleotides are preferably a contiguous part of SEQ ID NOs 1 to 22, i.e. the nucleotides are consecutive in the respective SEQ ID NO.

The molecule according to item (a) is also preferably a "shRNA". shRNA preferably utilizes the U6 promoter for its expression. The shRNA according to the invention comprises or consists a sequence which is with increasing preference complementary to at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, or all 23 nucleotides of SEQ ID NOs 1 to 22. These at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, or at least 21 nucleotides are preferably a contiguous part of SEQ ID NOs 1 to 22, i.e. the nucleotides are consecutive in the respective SEQ ID NO.

An molecule according to item (b) of the above preferred embodiment of the invention is capable of interacting with, more specifically hybridizing with the target IncRNA. By formation of the hybrid the function of the IncRNA is reduced or blocked. Standard methods relating to such antisense technology have been described (see, e.g., Melani et al., Cancer Res. (1991) 51 :2897-2901). The term "antisense molecule" in accordance with the present invention thus relates to a nucleic acid molecule, preferably a RNA molecule, that has a base sequence complementary to a given IncRNA, i.e. the "sense" sequence. A particularly preferred example of the molecule according to item (b) is an Endoribonuclease- prepared siRNA (esiRNA).

The sequence identity of the antisense molecule according to item (b) to an IncRNA selected from SEQ ID NOs 1 to 22 is with increasing preference at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99% and 100%. Means and methods for determining sequence identity are known in the art. Preferably, the BLAST (Basic Local Alignment Search Tool) program is used for determining the sequence identity with regard to one or more IncRNAs selected from SEQ ID NOs 1 to 22. Antisense molecules, siRNAs and shRNAs of the present invention are preferably chemically synthesized using appropriately protected ribonucleoside phosphoramidites and a conventional RNA synthesizer. Suppliers of RNA synthesis reagents include Proligo (Hamburg, Germany), Dharmacon Research (Lafayette, CO, USA), Pierce Chemical (part of Perbio Science, Rockford, IL , USA), Glen Research (Sterling, VA, USA), ChemGenes (Ashland, MA, USA), and Cruachem (Glasgow, UK).

As discussed in greater detail herein above, the ability of antisense molecules, siRNA, and shRNA to potently, but reversibly, silence IncRNA and genes in vivo makes these molecules particularly well suited for use in the pharmaceutical composition of the invention. The antisense molecules, siRNAs, shRNAs may comprise modified nucleotides such as locked nucleic acids (LNAs). LNA nucleotides can be mixed with DNA or RNA residues in the oligonucleotide whenever desired. Particulary, preferred example of siRNAs is GapmeR (LNA™ GapmeRs (Exiqon)). The GapmeRs are preferably 14-16 nucleotides in length and are optionally fully phosphorothioated.

Examples of suitable expression vector which may be used in connection with item (d) of the above-preferred embodiment have been detailed herein above.

In accordance with a preferred embodiment of the third and fourth aspect of the invention the compound as defined in (ii) is an aptamer, a ribozyme, an antibody, a protein drug, or a small molecule inhibitor.

The aptamer, ribozyme, antibody, protein drug, or small molecule inhibitor of this embodiment specifically bind to one or more IncRNA selected from SEQ ID NOs 1 to 22, thereby inhibiting the activity of one or more IncRNA selected from SEQ ID NOs 1 to 22. The terms "aptamer", "ribozyme", "antibody", "protein drug", and "small molecule inhibitor" have been defined herein above in connection with the first and second aspect of the invention and apply mutatis mutandis to the third and fourth aspect of the invention. The present invention relates in a fifth aspect to a method for diagnosing hypoxia in a patient, comprising (a) detecting the expression level of one or more IncRNAs selected from SEQ ID NOs 1 to 42, preferably selected from SEQ ID NOs 1 to 3 and 14 in a sample obtained from said patient; and (b) comparing said expression level of the one or more IncRNAs with the expression level of these one or more IncRNAs in a sample obtained from healthy subjects, wherein a greater than 2-fold upregulation of one or more IncRNAs selected from SEQ ID NOs 1 to 22; and/or a greater than 2-fold downregulation of one or more IncRNAs selected from SEQ ID NOs 23 to 42 is indicative for hypoxia in the patient.

The method according to the fifth aspect of the invention may also encompass detecting and comparing the expression level of one or more IncRNAs being with increased preference at least 90%, at least 92%, at least 94%, at least 96%, at least 98%, at least 99%, and at least 99.5% identical to any one of SEQ ID NOs 1 to 22 and/or 23 to 42. Means and methods for determining sequence identity are known in the art. Preferably, the BLAST (Basic Local Alignment Search Tool) program is used for determining the sequence identity with regard to one or more IncRNAs selected from SEQ ID NOs 1 to 22 and/or 23 to 42. The method according to the fifth aspect of the invention may furthermore encompass detecting and comparing the expression level of one or more IncRNAs differing with increasing preference by no more than 10, such as 5, 4, 3, 2 or 1 nucleotide(s) from any one of SEQ ID NOs 1 to 22 and/or 23 to 42. The nucleotide differences may be the addition, deletion and/or substitution of nucleotide(s). The sequences the expression of which is compared, while being homologous, may also differ from each other with increasing preference by no more than 10, such as 5, 4, 3, 2 or 1 nucleotide(s).

The term "hypoxia" designates a pathological condition in which the body or a region of the body is deprived of an adequate oxygen supply. Hypoxia may be classified as either generalized, affecting the whole body, or local, affecting a region of the body. Hypoxia occurs in tumors. Tumor hypoxia is the situation where tumor cells have been deprived of oxygen. As a tumor grows, it rapidly outgrows its blood supply, leaving portions of the tumor with regions where the oxygen concentration is significantly lower than in healthy tissues. Ischemia, meaning insufficient blood flow to a tissue, also results in hypoxia. Hence, the above method is preferably used for diagnosing tumor hypoxia and/or ischemic hypoxia.

The term "sample" designates a tissue sample or a body fluid sample. The body fluid sample is preferably selected from blood, serum, plasma, urine, salvia, amniotic fluid, cerebrospinal fluid and lymph. The tissue sample is preferably an organ sample, such as a heart, liver or kidney sample. Because in the examples provided herein below the expression of the IncRNA has been detected in Human Umbilical Vein Endothelial Cells (HUVECs) the sample preferably comprises or consists of endothelial cells, or at least comprises the IncRNAs from endothelial cells. More preferably the sample comprises or consists of human endothelial cells, or at least comprises the IncRNAs from human endothelial cells. The sample comprises in general the IncRNA of whole cells but may also only comprise the IncRNA of the cytoplasmic compartment or the nuclear compartment. The examples herein below exemplarily show for the IncRNAs DSCAM-1 and PLAC1-1 that IncRNA expression can be detected in the cytoplasmic compartment as well as the nuclear compartment (see Figures 3 and 1 1 ). In case the sample only comprises the IncRNAs of a particular cellular compartment, the cytoplasmic compartment is preferred. As far as the method is applied to a body fluid sample it is to be understood that the expression level of an IncRNA corresponds to the concentration of the IncRNA, because IncRNAs are not directly expressed in the body fluid but secreted from the cells, said cells expressing the IncRNAs, into the body fluids.

The "patient" or "subject" referred to herein is human.

The term "detecting the expression level of IncRNA" means determining the amount or yield of the IncRNA. The IncRNAs are initially expressed within a cell. It was found in accordance with the present invention that the IncRNAs of SEQ ID NOs 1 to 22 and/or 23 to 42 can be detected in the sample of a patient, in particular in various tissues including heart tissue. An IncRNA being "expressed in a sample" is therefore a IncRNA whose expression level can be detected in the sample by means and methods being further detailed herein below. An ncRNA is upregulated in a test sample if the amount or yield of the ncRNA is significantly greater as compared to the amount or yield of the corresponding ncRNA in a control sample. Likewise, an ncRNA is downregulated in a test sample if the amount or yield of the ncRNA is significantly less as compared to the amount or yield of the corresponding ncRNA in a control sample. In this context the term "corresponding ncRNA" means, for example, that the expression level of the IncRNA of SEQ ID NO: 1 in the test sample is compared to the expression level of the IncRNA of SEQ ID NO: 1 in the control sample, or likewise that the expression level of the IncRNA of SEQ ID NO: 2 in the test sample is compared to the expression level of the IncRNA of SEQ ID NO: 2 in the control sample. This applies mutatis mutandis for scenarios where the expression of more than one IncRNA selected from SEQ ID NOs 1 to 22 and/or 23 to 42 is determined. For instance, if the expression level of all IncRNAs of SEQ ID 1 to 22 and/or 23 to 42 is determined in the test sample it is compared to the expression level of ail IncRNAs of SEQ ID NOs 1 to 22 and/or 23 to 42 in the control sample.

The expression level in the samples can be quantified by any suitable means and methods available from the art. In general relative and absolute quantification means and methods can be used. In absolute quantification no known standards or controls are needed. The expression level can be directly quantified. As well-known in the art, absolute quantification may rely on a predetermined standard curve. In relative quantification the expression level is quantified relative to a reference (such as known control expressions levels). Also in the absence of controls, one can relatively quantify the expression level when comparing e.g. fluorescence intensities.

Methods to assess RNA concentration may, for example, comprise measuring the fluorescence intensity of dyes that bind to nucleic acids and selectively fluorescence when bound. Such methods comprise a reverse transcription reaction and the production of cDNA, wherein the amount of the cDNA is determined thereby indirectly determining the amount of the RNA. The fluorescent-based method is particularly useful for cases where the RNA concentration is too low to accurately assess some with spectrophotometry and/or in cases where contaminants absorbing at 260nm make accurate quantification by spectrophotometry difficult or impossible.

When comparing the expression level of the one or more IncRNAs between different samples reliability of the comparison is preferably improved by including an invariant endogenous control (expression of a reference gene) to correct for potential sample to sample variations. Such normalization with respect to an invariant endogenous control is routinely performed in the art. For example, means and methods for expression level normalization, e.g. in real-time RT-PCR (see, for example, Bustin, Journal of Molecular Endocrinology, (2002) 29, 23-39) or micro-array expression analysis (see, for example, Calza and Balwitan, Methods Mol Biol. 2010;673:37-52) are well-established. Also methods for normalization of the expression levels of small RNA sequences are established (see, for example, Mestdagh et al. (2009) Genome Biol.; 10(6):R64). In case RT-PCR or a micro-array is used to determine the expression levels in accordance with the present invention, the expression levels are preferably normalized to a spiked-in RNA (see, for example, McCormick et al. (201 1 ), Silence, 2:2). Known amounts of a spiked-in RNA are mixed with the sample during preparation. More preferably the RNA is externally spiked-in to plasma and/or serum before the RNA isolation process is carried out, in which case the samples are plasma and/or serum. The spiked-in RNA technology is well- known and commercial kits are available from a number of manufacturers. The spiked-in RNA is preferably a spiked-in C. elegans RNA. As evident from the examples herein below, the deregulation of the levels of one or more IncRNAs selected from 1 to 22 and/or 23 to 42, are indicative for hypoxia. Thus, determining the expression levels of one or more IncRNAs selected from 1 to 22 and/or 23 to 42 can be expected to be of prognostic value for diagnosing a hypoxia in a patient. The IncRNAs selected from 1 to 22 and/or 23 to 42 may be combined with further diagnostic markers for hypoxia in order to enhance the confidentially of the diagnostic method. High-expression level of the IncRNAs selected from 1 to 22 and low expression level of the IncRNAs selected from 23 to 42 is indicative for hypoxia.

In the examples herein below the primer sequences of SEQ ID NOs 45 to 52 were employed in order to detect the expression level of the three different isoforms of DSCA -1 (SEQ ID NOs 1 to 3) and PLAC1-1 , wherein the uneven numbers are forward primers and the even numbers are reverse primers. Consecutive numbers, such as SEQ ID NOs 45 and 46, SEQ ID NOs 47 and 48 etc. are a primer pair. As defined herein, SEQ ID NO: 1 is the isoform 3, SEQ ID NO: 2 is the isoform 2, and SEQ ID NO: 3 is the isoform 1 of DSCAM-1. The primer pair of SEQ ID NOs 45/46 is for the detection of the expression level of isoform 1 of DSCAM-1 , SEQ ID NOs 47/48 is for isoform-2 of DSCAM-1 , and SEQ ID NOs 49/50 is for isoform-3 of DSCAM-1. The primer pair of SEQ ID NOs 51/52 is for the detection of the expression level of the IncRNA PLAC1-1 (SEQ ID NO: 14).

One or more of these primer pairs are preferably used in the diagnostic method according to the third aspect of the invention. One or more of these primer pairs are likewise preferably incorporated into the kit of the invention being described herein below.

The greater than 2-fold downregulation is with increasing preference greater than 3-fold downregulation, greater than 4-fold downregulation, greater than 5-fold downregulation, greater than 6-fold downregulation, greater than 7-fold downregulation and greater than 8-fold downregulation. Likewise the greater than 2-fold upregulation is with increasing preference greater than 3-fold upregulation, greater than 4-fold upregulation, greater than 5-fold upregulation, greater than 6-fold upregulation, greater than 7-fold upregulation and greater than 8-fold upregulation. The higher thresholds for the up- and downregulation may increase the reliability of the method of the third aspect of the invention.

In accordance with a preferred embodiment of the fifth aspect of the invention the sample is a blood sample or blood-derived sample. The blood-derived sample is preferably plasma or serum.

In accordance with another preferred embodiment of the fifth aspect of the invention the sample is a tissue sample.The tissue sample comprises preferably heart tissue, and more preferably ECs of the heart.

In accordance with a further preferred embodiment of the fifth aspect of the invention the detection of the expression level of the one or more IncRNAs comprises (a) quantitative PCR, preferably quantitative real time PCR, or (b) a template/RNA amplification method followed by determining the expression level of the one or more IncRNAs using a fluorescence- or luminescence-based quantification method.

In quantitative PCR (qPCR), the amount of amplified product is linked to fluorescence intensity using a fluorescent reporter molecule. The point at which the fluorescent signal is measured in order to calculate the initial template quantity can either be at the end of the reaction (endpoint semi-quantitative PCR) or while the amplification is still progressing (real-time qPCR).

In endpoint semi-quantitative PCR, fluorescence data are collected after the amplification reaction has been completed, usually after 30-40 cycles, and this final fluorescence is used to back-calculate the amount of template present prior to PCR. The more sensitive and reproducible method of real-time qPCR measures the fluorescence at each cycle as the amplification progresses. This allows quantification of the template to be based on the fluorescence signal during the exponential phase of amplification, before limiting reagents, accumulation of inhibitors, or inactivation of the polymerase have started to have an effect on the efficiency of amplification. Fluorescence readings at these earlier cycles of the reaction will measure the amplified template quantity where the reaction is much more reproducible from sample to sample than at the endpoint.

A non-limiting example of a template/RNA amplification method followed by determining the expression level of the one or more IncRNAs using a fluorescence- or luminescence-based quantification method is a method combining transcription mediated amplification (TMA) and a hybridization protection assay (HPA). In more detail, such a method may comprise hybridizing one or more oligonucleotides ("capture oligonucleotides") that are complementary to any of SEQ ID NOs 1 to 22 or 23 to 42. In case two or more of SEQ ID NOs 1 to 22 and 23 to 42 are targeted, a separate capture oligonucleotides is used for each sequence selected from 1 to 22 and 23 to 42. The hybridized target sequences are then captured onto magnetic microparticles that are separated from the sample in a magnetic field. Wash steps may be utilized to remove extraneous components. Target amplification typically occurs via TMA, which is a transcription-based nucleic acid amplification method that utilizes two enzymes, Moloney murine leukemia virus (MMLV) reverse transcriptase and T7 RNA polymerase. A unique set of primers is used for each target sequence selected from 1 to 22 and 23 to 42. The reverse transcriptase is used to generate a DNA copy (containing a promoter sequence for T7 RNA polymerase) of the target sequence. T7 RNA polymerase produces multiple copies of RNA amplicon from the DNA copy. Detection of IncRNA expression level is achieved by HPA using single-stranded, chemiluminescent-labeled nucleic acid probes that are complementary to the one or more amplicon. Preferably, distinguishably labelled probes are used for each target amplicon. The labeled nucleic acid probes hybridize specifically to the amplicon. A "selection reagent" then differentiates between hybridized and unhybridized probes by inactivating the label on unhybridized probes. During the detection step, the chemiluminescent signal produced by the hybridized probe is measured in a luminometer and is reported as "Relative Light Units" (RLU), thereby quantifying the IncRNA expression level.

In accordance with a still further preferred embodiment of the fifth aspect of the invention the method comprises prior to the detection of the expression level of the long non-coding RNA a pre-amplification step of the RNA within the test patient's sample and/or the control patient's sample.

Performing a pre-amplification step is of particular advantage in case only a low amount of (test and/or control) sample is available. The pre-amplification step allows increasing the amount of RNA within the sample before proceeding to the analysis of the expression level. Means and methods for the pre-amplification of RNA are well known in the art (see, e.g., Vermeulen et al (2009) BMC Res Notes., 2:235). In case both the RNA in the test and control sample is pre-amplified preferably the same method for the pre-amplification step is used such that the relative amount of RNA of the test sample as compared to the control sample is maintained. In case only the RNA of the test or control sample is pre-amplified or the two RNA samples are pre-amplified by different methods, the expression level data may have to be normalized for pre-amplification step; see, e.g. Mestdagh et al. (2009), Genome Biology 2009, 10:R64.

The present invention relates in a sixth aspect to a kit for diagnosing hypoxia in a patient, said kit comprising means for the detection of the expression level of one or more IncRNAs selected from SEQ ID NOs 1 to 42, preferably selected from SEQ ID NOs 1 to 3 and 14 and instructions how to use the kit.

The instructions how to use the kit preferably inform inter alia that high-expression level of the IncRNAs selected from 1 to 22 and low expression level of the IncRNAs selected from 23 to 42 is indicative for hypoxia. The means for the detection of the expression level of one or more IncRNAs selected from SEQ ID NOs 1 to 22 and 23 to 42 are preferably the means required for (i) a quantitative PCR, preferably quantitative real time PCR, or (ii) a template/RNA amplification method followed by determining the expression level of the one or more IncRNAs using a fluorescence- or luminescence-based quantification method. These means have been further detailed herein above in connection with the fifth aspect of the invention, and may be comprised in the kit. Hence, the means preferably comprise oligonucleotides, such as fluorescent hybridization probes or primers, which specifically hybridize to one or more IncRNAs selected from SEQ ID NOs SEQ ID NOs 1 to 22 and 23 to 42. Additional ingredients of the kits may be florescent or luminescent dyes, preferably coupled to said oligonucleotides. Also, additional ingredients of the kits may be enzymes, such as a reverse transcriptase and/or a polymerase.

In accordance with the kit of the invention the means for the detection of the expression level of one or more IncRNAs selected from SEQ ID NOs SEQ ID NOs 1 to 22 and 23 to 42 preferably comprise means for the detection of the IncRNA of SEQ ID NOs 1 to 3.

The various components of the kit may be packaged in one or more containers such as one or more vials. The vials may, in addition to the components, comprise preservatives or buffers for storage.

In accordance with a preferred embodiment of the sixth aspect of the invention, the means are primer pairs used for the specific detection of the expression level of one or more IncRNAs selected from SEQ ID NOs SEQ ID NOs 1 to 22 and 23 to 42. In accordance with a preferred embodiment of all six aspects of the invention the one or more IncRNAs are at least 3 IncRNAs, and preferably at least 5 IncRNAs.

Employing at least 3 IncRNAs, preferably at least 5 IncRNAs, more preferably at least 10 IncRNAs, even more preferably at least 20 IncRNAs and most preferably all IncRNAs of SEQ ID NOs 1 to 22 and 23 to 42 will additionally increase the effectivity of the pharmaceutical compositions, medical uses, methods and kits of the invention. Employing these numbers of IncRNAs may balance potential differences associated with particular compounds, probes or methods used in connection with the methods and kits of the invention. In the pharmaceutical compositions and medical uses of the invention these numbers of IncRNAs may increase the beneficial effect for the subject to be treated. In accordance with a preferred embodiment of all six aspects of the invention the one or more IncRNAs is or comprises the IncRNA of SEQ ID NO: 1.

As discussed herein above SEQ ID NO: 1 is the IncRNA DSCA -1 isoform 3. The pro- angiogenic nature of IncRNA DSCAM-1 isoform 3 is demonstrated in the example herein by expression profiling and over-/under-expression experiments in HUVEC cells.

The figures show. Figure 1 : The three isoforms of the IncRNA DSCAM-1 (also designated herein LINC00323 or Inc323 or HSLINCR).

Figure 2: (A) Differential IncRNA expression after hypoxia in HUVECs (RNA-sequencing data). (B) HUVECs treated with VEGF (50 ng/ml) for 24 h. N = 3.

Figure 3: Validation of NCode array data for lnc-DSCAM-1 (LINC00323) in fractionated RNA.

Figure 4: Tissue distribution of lnc-DSCAM-1 (LINC00323). Figure 5: siRNA against lnc-DSCAM-1 isoform 3 (HSLINCR-003, SEQ ID NO: 1 ) impairs HUVEC proliferation.

Figure 6: Downstream signalling triggered by loss of lnc-DSCAM-1 isoform 3 (HSLINCR-003). Figure 7: Effect of lnc-DSCAM-1 isoform 3 (HSLINCR-003) knockdown on capillary growth.

Figure 8: Overexpression of lnc-DSCAM-1 , isoforms 1 and 3 (Iinc323-1 and linc323-3) in the human endothelial cell line Ea.Hy926. Figure 9: Angiogenic gene expression pattern upon overexpression of lnc-DSCAM-1 isoform 3 (Nnc323-3) in the human endothelial cell line Ea.Hy926.

Figure 10: Endothelial IncRNA lnc-DSCAM-1 isoform 3 (LINC00323-003; SEQ ID NO: 1 ) expression is crucial for endothelial cellular function. HUVECs treated with VEGF (50 ng/ml) for 24 h. N = 3. Figure 11 : Validation of the IncRNA lnc-PLAC1-1 (SEQ ID NO: 14) in subcellular RNA (total, nuclear, cytoplasmic) from HUVECs subjected to normoxia and hypoxia. Fold-change of normoxia is plotted. N = 3/4. * = p < 0.05, * * = p < 0.01 , ** * = p < 0.001 Figure 12: Endothelial IncRNA lnc-PLAC1-1 (SEQ ID NO: 14) is transcriptionally regulated with miR-503 after hypoxia and its loss deteriorates HUVEC function. (A) Genomic localization of lnc-PLAC1-1 , adapted from NCBI Map Viewer. (B) Primary transcript of miR-503 (pri-miR- 503) and processed, mature miR-503 are upregulated after 24 h hypoxia in HUVECs determined by qRT-PCR. N = 4. (C) GapmeR-mediated knockdown of the IncRNA lnc-PLAC1- 1 efficiently lowers endogenous expression level in HUVECs detected by qRT-PCR. N = 3. (D) Loss of lnc-PLAC1-1 causes impairment in BrdU incorporation rate monitored by ELISA. N = 3. (E) Scratch wound closure is impaired when the IncRNA lnc-PLAC1-1 is silenced in HUVEC. N = 3. (F) Increase in cell cycle inhibitor p21 expression detected by qRT-PCR. N = 3. (G) GapmeR against the IncRNA lnc-PLAC1-1 represses GATA2 on protein level. N = 4. (H) Migration index is improved in transgenic Ea.Hy926 cells overexpressing LINC00323-003 and lnc-PLAC1-1 as compared to the control. N = 5. * = p < 0.05, * * = p < 0.01 , *** = p < 0.001

The examples illustrate the invention. Example 1 - Identification of hypoxia-sensitive IncRNAs

It was studied if long non-coding RNAs (IncRNAs) would be differentially expressed after hypoxic treatment in human umbilical vein endothelial cells (HUVECs). For RNA deep sequencing analysis total RNA derived from human umbilical vein endothelial cells (HUVECs) that underwent in vitro cultivation under normoxic or hypoxic (0.2% 0 2 ) conditions for 24 h was generated. Taking into account biological variations within the groups, N = 3 per group for HUVECs were employed.

Hypoxia-sensitive non-coding RNA (ncRNA) expression was detected via two approaches; (a) microarray- and (b) RNA-sequencing (RNA-seq) techniques.

NCode (Invitrogen) microarray was performed to analyze differential expression of noncoding RNA during hypoxia of human umbilical vein endothelial cells (HUVECs). Evaluation of this data revealed appearance of hypoxia-sensitive IncRNAs (Fig. 2A). These IncRNAs are shown in Table 1 and SEQ ID NOs 1 to 42 of the application as filed. The hypoxia-sensitive IncRNAs are significantly deregulated under hypoxic conditions. SEQ ID NOs 1 to 22 were found to be upregulated, while EQ ID NOs 1 to 22 were found to be downregulated.

Table 1 : Hypoxia-sensitive IncRNAs

Inc NA name feature length base Mean !los2 Fold Char D value padi lnc-WDR74-l » 3756; 1175,3957! -1,4277703 7,94E-94 5,67E-91 lnc-SAMD14-2 4290 1266,7685: -0,9997163 7,25E-96: 6,21E-93 lnc-AKRlC2-4 4450 1561,2652) -0,9389733; 3,14E-69 : l,28E-66 lnc-SEBOX-2 2662 1722,7503 -0,9144215: 4,12E-71 ; l,96E-68 l c-C9orf69-l 3389 1462,7341; -0,9012412 l,20E-44 2,45E-42 lnc-SERPINCl-1 : 3631i 1809,5211 -0,7581272) 5,52E-19 3,94E-17 lnc-GJAlO-3 9544; 1416,5181; -0,7563964, l,03E-69 4,64E-67 lnc-ANKRD12-l 1050; 1229,3493! -0,7283248' l,89E-22; l,60E-20 lnc- RPS25-l 5131 4034,2832; -0,5751249 2,04E-88 l,34E-85 lnc-B3GAT2-3 66091 1065,9363; -0,4584332 7,04E-25 6,34E-23 lnc-C6orfl46-3 7070i 1845,0425 ^ 0,4459623! 2,89E-30 3,75E-28 lnc-DLKl-4 13388i 9850,0601; 0,5521516 2,80E-122 3,42E-119 lnc-IDS-1 4581; 1341,6273 0,563006, 9,63E-50 2,84E-47 lnc-ZCCHC7-2 4375; 1363,4439; 0,6099449 2,86E-4i; 5,10E-39 lnc-AKl-1 1160 1363,16 0,8456338; l,10E-56; 3,92E-54 lnc-C14orfl66B-l 721 1312,6298 0,9946821 2,96E-42 5,64E-40 lnc-LCN6-l ; 4345 1649,2145 1,0725203 5,15E-35 7,36E-33 lnc-FNl-3 5628 ' 7159,3619 : 1,1548762 0 0

1678 1049,2302 ' 2,141139 1,04Ε-261 2,96Ε-258

533; 439,7136 ! 5,09995 ; 1,25Ε-158 2,68Ε-155

2321 136,0999 4,0410731 , 2,19Ε-55 7,46Ε-53;

7232 : 244,68909 ; 3,0583206 6,07Ε-79 3,71Ε-76

11246! 109,93138 ! 3,033384 ! 5,37Ε-59 2,00Ε-56

2138 : 601,59313 s 2,7153192 3,54Ε-128 5,06Ε-125

1965 702,82234 : 2,655259 1,06Ε-274 4,54Ε-271

4326ί 113,11347 2,6471848 3,13Ε-47 7,89Ε-45

3101 114,24598 1 2,4725645 3,95Ε-47 9,67Ε-45

2301 ; 158,884 , 2,340892 , 2,08Ε-51 6,35Ε-49

2095 292,66698 ! 2,3222268 2,29Ε-102 2,18Ε-99

1571' 88,112301 ! -1,815099 ! 1,88Ε-27 ! 2,13Ε-25

777 : 43,331816 -1,8986651 : 7,10Ε-13 3,09Ε-11

13458 ! 91,303344 -1,9138273 9,79Ε-18 : 6,40Ε-16

2449 : 71,845142 -1,9563842 3,10Ε-27 3,45Ε-25

2428! 397,2283 ; -1,9701526 ! 7,03Ε-116 7,52Ε-113

1623 276,22509 ; -2,3054837 3,86Ε-94 , 3,00Ε-91

1753 ! 77,561264 -2,4417613 4,24Ε-29 5,18Ε-27

4248! 494,95654 ! -2,5229731 : 2,40Ε-138 4,11Ε-135

645 88,433128 -2,8220783 3,51Ε-39 5,76Ε-37

557 : 57,6028! -3,4966215 2,18Ε-37 ! 3,33Ε-35

2432 29,239401 ; 2,1091475 , 6,09Ε-12 ! 2,46Ε-10

Moreover, RNA-seq data identified 774 (404 up, 371 down) significantly deregulated IncRNAs (Fig. 2B). In order to obtain the RNA-seq data 2 Mg of total RNA - isolated from HUVEC cells cultured under normoxic or hypoxic conditions, respectively for 24 h - was subjected to RNA- Seq analysis (N = 3). After extracting the total RNA from the samples, rRNA was removed from the total RNA. By using the fragmentation buffer, the remaining RNA was fragmented into short fragments. The first strand cDNA was synthesized by random hexamer-primer using the remaining RNA fragments as templates. Buffer, dNTPs, RNase H and DNA polymerase I were added to synthesize the second strand. The double strand cDNA was purified with QiaQuick PCR extraction kit and washed with EB buffer for end repair and poly (A) addition. Finally, sequencing adaptors were ligated to the fragments. The fragments were purified by agarose gel electrophoresis and enriched by PCR amplification. The library products were sequenced via lllumina HiSeqTM 2000.

The log2-fold expression changes derived from the two different technologies - NCode (Invitrogen) microarray data and RNA-seq data - were significantly correlated both for IncRNAs (Pearson correlation coefficient: 0.43; p-value < 2.2e-16) and for protein-coding genes (Pearson correlation coefficient: 0.74; p-value < 2.2e-16). RNA-Seq also confirmed enhanced expression of LINC00323 (lnc-DSCAM-1 ) under hypoxia. In addition, from the RNA- Seq data another hypoxia-sensitive IncRNA lnc-PLAC1-1 (SEQ ID NO: 14 or MIR503HG-002) was identified based on the following critera: (a) minimal 4-fold upregulation, (b) base mean > 1000, and c) intergenic annotation. Based on these stringent criteria lnc-PLAC1-1 was identified as high propriety candidate. The subsequent validation experiments being described in the following examples 2 and 3 confirm the hypoxia-dependent upregulation of the two microarray- or RNA-seq derived IncRNAs LINC00323 and lnc-PLAC1-1.

Example 2 - Functional experiments followed to decipher endothelial function of the IncRNA DSCAM-1 (LINC00323 or HSLINCR)

As can be taken from Table 1 the long intergenic noncoding RNA (LINCRNA) 323 was upregulated in the microarray data. DSCAM-1 has three transcript variants (lncRNA323-1 (SEQ ID NO: 3), lncRNA323-2 (SEQ ID NO: 2), and lncRNA323-3 (SEQ ID NO: 1 )).

HUVECs were cultured under normoxic or hypoxic (0.2% 02, 24 h) conditions and total RNA was analyzed by microarray and next generation RNA-sequencing analysis (see Example 1 ). Total RNA was fractionated to a cytoplasmic and nuclear fraction and expression levels of hypoxiasensitive IncRNAs were validated via qPCR. Loss-of-function-experiments using siRNA against specific IncRNAs was applied to determine proliferation, apoptosis, capillary tube formation and gene or protein expression analysis. Gain-of-function-approaches via lentiviral delivery was applied to characterize functional IncRNA overexpression.

Example 2.1 - qPCR analysis of endothelial lnc-DSCAM-1 transcripts

Endothelial lnc-DSCAM-1 transcripts were validated to be hypoxia-sensitive via qPCR analysis. Of note, endothelial lnc-DSCAM-1 transcript expression was upregulated in the cytoplasmic RNA fraction after hypoxia. N=3 experiments per group (Fig 3). Example 2.2 - Tissue distribution of lnc-DSCAM-1 transcripts

Human lnc-DSCAM-1 transcript variants were detected in several organ tissues. DSCAM-1 is ubiquitously expressed with highest expression in vascularized tissue such as kidney or lung (Fig 4). Of note, DSCAM-1 transcript 2 (SEQ ID NO: 2) is not expressed in every tissue. Example 2.3 - siRNA against lnc-DSCAM-1 isoform 3 lnc-DSCAM-1 isoform 3 (LINC00323-003, HSLINCR-003, SEQ ID NO: 1 ) knockdown deteriorates HUVEC function by inhibiting proliferative pathways. (Fig 5A) WST1-acitivity is reduced in HSLINCR siRNA-transfected HUVECs. (Fig 5B) BrdU incorporation rate is impaired in DSCAM-1 isoform 3-deficient HUVECs. (Fig5 C) HSLINCR (LINC00323-003) loss has no effect on apoptosis rate. N = 3 experiments per group. * = p < 0.05, *** = p < 0.001.

Example 2.4 - Downstream signalling triggered by loss of lnc-DSCAM- .

(Fig 6A) Growth factor-related ERK Akt signalling is decreased by low lnc-DSCAM-1 (LINC00323-003, HSLINCR) expression contributing to the defective functional phenotype. Representative Western Blot is shown. (Fig 6B) Cell cycle inhibitors p21 and p27 are upregulated after siRNA against DSCAM-1 (LINC00323-003). (Fig 6C) Pro-angiogenic GATA2 and SIRT1 are repressed after loss of DSCAM-1 (LINC00323-003). N = 3/4 experiments per group. * = p < 0.05, ** = p < 0.01

Example 2.5 - lnc-DSCAM-1 knockdown and capillary growth Lnc-DSCAM-1 isoform 3 (LINC00323-003, HSLINCR-003) knockdown inhibits capillary growth (Fig 7). (Fig 7A) Endogenous modulation of HSLINCR (LINC00323-003) triggers angiogenic defects in a matrigel-based tube forming assay. (Fig 7B) Pro- and anti-angiogenic cytokines VEGFB and THBS1 are affected by lnc-DSCAM-1 modulation. N = 3 experiments per group. * = p < 0.05, * * = p < 0.01

Example 2.6 - lnc-DSCAM-1 overexpression studies in human endothelial cell line Ea.Hy926 lnc-DSCAM-1 isoform 3(LINC00323-003) transcripts were cloned into a lentiviral backbone to generate viral particles for overexpression studies. qPCR analysis was performed to study lnc-DSCAM-1 (being a LINCRNA) overexpression. Fig 8 shows the lnc-DSCAM-1 , isoforms 1 and 3 expression rate in human endothelial cell line Ea.Hy926. Example 2.7 - Angiogenic gene expression pattern upon overexpression of lnc-DSCAM-1 , isoform 3

Angiogenic gene expression pattern in human endothelial cell line Ea.Hy926 constitutively overexpressing lnc-DSCAM-1 , isoform 3 in comparison to control cells.

Example 2.8 - VEGF-dependent induction of lnc-DSCAM-1 expression

VEGF treatment increased lnc-DSCAM-1 , isoform 3 (LINC00323-003) expression (Fig 10).

Example 3 - Functional experiments followed to decipher endothelial function of the IncRNA lnc-PLAC1-1 (MIR503HG-002 , NONHSAT138623 or NR_024607)

Example 3.1 - Hypoxia-dependent enhanced expression of lnc-PLAC1-1 in the nuclear and cytoplasmic compartment

Cell compartment-specific RNA analysis revealed hypoxia-dependent enhanced expression, of lnc-PLAC1-1 in both the nuclear and cytoplasmic compartment (Fig 1 1 ). High purity of subcellular compartment preparations was proven by screening expression of the nuclear enriched IncRNA XIST.

Example 3.2 - Endothelial functions of hypoxia-sensitive IncRNA lnc-PLAC1-1

The RNA-seq derived IncRNA PLAC1-1 is located at chromosome X, harbors a coding sequence of miR-503 and is adjacent to miR-424 (Fig 12A). Indeed, analysis of pri-miR-503 and mature miR-503 expression after hypoxia showed a similar increase of expression reflecting same regulatory mechanisms for transcriptional activation (Fig 12B). Both miR-503 and the IncRNA PLAC1-1 modulations had different effects on endothelial cell function; whereas transfection of miR-503 precursors resulted in negative effects on endothelial cell viability, knockdown of lnc-PLAC1-1 suppressed endothelial proliferation independent of alterations of apoptosis (Fig 12C and D). Inc-PLAC1-1 silencing via GapmeR technology led to reduced miR-503 expression. In contrast, transfection of miR-503 precursors did not alter Inc- PLAC1-1 expression suggesting dependency of miR-503 expression on lnc-PLAC1-1 but not wee versa. Migratory capacity of lnc-PLAC1-1 -deficient endothelial cells was additionally inhibited (Fig 12E). The proliferative and migratory functional defects in endothelial cells after lnc-PLAC1-1 silencing were paralleled by a strong upregulation of cell cycle inhibitor p21 (Fig 12F). Silencing of lnc-PLAC1-1 repressed expression of the key angiogenic factor GATA2 (Fig 3G).

Example 4 - Enhanced expression of pro-angiogenic IncRNAs improves migratory capacity

To study the role of enhanced IncRNA expression in endothelial cells, hypoxia-sensitive IncRNAs lnc-DSCAM-1 , isoform 3 (LINC00323-003) and lnc-PLAC1-1 were both overexpressed by generation of stable GFP-expressing endothelial cell lines (Ea.Hy926). The migration index is improved in transgenic Ea.Hy926 cells overexpressing LINC00323-003 and lnc-PLAC1-1 as compared to the control. Transgenic Ea.Hy926 constitutively overexpressing LINC00323-003 even revealed an significantly improved endothelial healing in the scratch wound assay presumably triggered by enhanced cyto-protective HMOX1 expression (Fig 12H).