Compositions and methods relating to lung specific genes

Information

  • Patent Application
  • 20050026211
  • Publication Number
    20050026211
  • Date Filed
    September 02, 2004
    19 years ago
  • Date Published
    February 03, 2005
    19 years ago
Abstract
The invention relates to LSG polypeptides, polynucleotides encoding the polypeptides, methods for producing the polypeptides, in particular by expressing the polynucleotides, and agonists and antagonists of the polypeptides. The invention further relates to methods for utilizing such polynucleotides, polypeptides, agonists and antagonists for applications, which relate, in part, to research, diagnostic and clinical arts.
Description
FIELD OF THE INVENTION

The present invention relates to newly identified nucleic acids and polypeptides present in normal and neoplastic lung cells, including fragments, variants and derivatives of the nucleic acids and polypeptides. The present invention also relates to antibodies to the polypeptides of the invention, as well as agonists and antagonists of the polypeptides of the invention. The invention also relates to compositions comprising the nucleic acids, polypeptides, antibodies, variants, derivatives, agonists and antagonists of the invention and methods for the use of these compositions. These uses include identifying, diagnosing, monitoring, staging, imaging and treating lung cancer and non-cancerous disease states in lung, identifying lung tissue, monitoring and modifying lung embryonic development and differentiation, and identifying and/or designing agonists and antagonists of polypeptides of the invention. The uses also include gene therapy, production of transgenic animals and cells, and production of engineered lung tissue for treatment and research.


BACKGROUND OF THE INVENTION

Throughout the last hundred years, the incidence of lung cancer has steadily increased, so much so that now in many countries, it is the most common cancers. In fact, lung cancer is the second most prevalent type of cancer for both men and women in the United States and is the most common cause of cancer death in both sexes. Lung cancer deaths have increased ten-fold in both men and women since 1930, primarily due to an increase in cigarette smoking, but also due to an increased exposure to arsenic, asbestos, chromates, chloromethyl ethers, nickel, polycyclic aromatic hydrocarbons and other agents. See Scott, Lung Cancer: A Guide to Diagnosis and Treatment, Addicus Books (2000) and Alberg et al., in Kane et al. (eds.) Biology of Lung Cancer, pp. 11-52, Marcel Dekker, Inc. (1998). Lung cancer may result from a primary tumor originating in the lung or a secondary tumor which has spread from another organ such as the bowel or breast. Although there are over a dozen types of lung cancer, over 90% fall into two categories: small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC). See Scott, supra. About 20-25% of all lung cancers are characterized as SCLC, while 70-80% are diagnosed as NSCLC. Id. A rare type of lung cancer is mesothelioma, which is generally caused by exposure to asbestos, and which affects the pleura of the lung. Lung cancer is usually diagnosed or screened for by chest x-ray, CAT scans, PET scans, or by sputum cytology. A diagnosis of lung cancer is usually confirmed by biopsy of the tissue. Id.


SCLC tumors are highly metastatic and grow quickly. By the time a patient has been diagnosed with SCLC, the cancer has usually already spread to other parts of the body, including lymph nodes, adrenals, liver, bone, brain and bone marrow. See Scott, supra; Van Houtte et al. (eds.), Progress and Perspective in the Treatment of Lung Cancer, Springer-Verlag (1999). Because the disease has usually spread to such an extent that surgery is not an option, the current treatment of choice is chemotherapy plus chest irradiation. See Van Houtte, supra. The stage of disease is a principal predictor of long-term survival. Less than 5% of patients with extensive disease that has spread beyond one lung and surrounding lymph nodes, live longer than two years. Id. However, the probability of five-year survival is three to four times higher if the disease is diagnosed and treated when it is still in a limited stage, i.e., not having spread beyond one lung. Id.


NSCLC is generally divided into three types: squamous cell carcinoma, adenocarcinoma and large cell carcinoma. Both squamous cell cancer and adenocarcinoma develop from the cells that line the airways; however, adenocarcinoma develops from the goblet cells that produce mucus. Large cell lung cancer has been thus named-because the cells look large and rounded when viewed microscopically, and generally are considered relatively undifferentiated. See Yesner, Atlas of Lung Cancer, Lippincott-Raven (1998).


Secondary lung cancer is a cancer initiated elsewhere in the body that has spread to the lungs. Cancers that metastasize to the lung include, but are not limited to, breast cancer, melanoma, colon cancer and Hodgkin's lymphoma. Treatment for secondary lung cancer may depend upon the source of the original cancer. In other words, a lung cancer that originated from breast cancer may be more responsive to breast cancer treatments and a lung cancer that originated from the colon cancer may be more responsive to colon cancer treatments.


The stage of a cancer indicates how far it has spread and is an important indicator of the prognosis. In addition, staging is important because treatment is often decided according to the stage of a cancer. SCLC is divided into two stages: limited disease, i.e., cancer that can only be seen in one lung and in nearby lymph nodes; and extensive disease, i.e., cancer that has spread outside the lung to the chest or to other parts of the body. For most patients with SCLC, the disease has already progressed to lymph nodes or elsewhere in the body at the time of diagnosis. See Scott, supra. Even if spreading is not apparent on the scans, it is likely that some cancer cells may have spread away and traveled through the bloodstream or lymph system. In general, chemotherapy with or without radiotherapy is often the preferred treatment. The initial scans and tests done at first will be used later to see how well a patient is responding to treatment.


In contrast, non-small cell cancer may be divided into four stages. Stage I is highly localized cancer with no cancer in the lymph nodes. Stage II cancer has spread to the lymph nodes at the top of the affected lung. Stage III cancer has spread near to where the cancer started. This can be to the chest wall, the covering of the lung (pleura), the middle of the chest (mediastinum) or other lymph nodes. Stage IV cancer has spread to another part of the body. Stage I-III cancer is usually treated with surgery, with or without chemotherapy. Stage IV cancer is usually treated with chemotherapy and/or palliative care.


A number of chromosomal and genetic abnormalities have been observed in lung cancer. In NSCLC, chromosomal aberrations have been described on 3p, 9p, 11p, 15p and 17p, and chromosomal deletions have been seen on chromosomes 7, 11, 13 and 19. See Skarin (ed.), Multimodality Treatment of Lung Cancer, Marcel Dekker, Inc. (2000); Gemmill et al., pp. 465-502, in Kane, supra; Bailey-Wilson et al., pp. 53-98, in Kane, supra. Chromosomal abnormalities have been described on 1p, 3p, 5q, 6q, 8q, 13q and 17p in SCLC. Id. In addition, the loss of the short arm of chromosome 3p has also been seen in greater than 90% of SCLC tumors and approximately 50% of NSCLC tumors. Id.


A number of oncogenes and tumor suppressor genes have been implicated in lung cancer. See Mabry, pp. 391-412, in Kane, supra and Sclafani et al., pp. 295-316, in Kane, supra. In both SCLC and NSCLC, the p53 tumor suppressor gene is mutated in over 50% of lung cancers. See Yesner, supra. Another tumor suppressor gene, FHIT, which is found on chromosome 3p, is mutated by tobacco smoke. Id.; Skarin, supra. In addition, more than 95% of SCLCs and approximately 20-60% of NSCLCs have an absent or abnormal retinoblastoma (Rb) protein, another tumor suppressor gene. The ras oncogene (particularly K-ras) is mutated in 20-30% of NSCLC specimens and the c-erbB2 oncogene is expressed in 18% of stage 2 NSCLC and 60% of stage 4 NSCLC specimens. See Van Houtte, supra. Other tumor suppressor genes that are found in a region of chromosome 9, specifically in the region of 9p21, are deleted in many cancer cells, including p16INK4A and p15INK4B. See Bailey-Wilson, supra; Sclafani et al., supra. These tumor suppressor genes may also be implicated in lung cancer pathogenesis.


In addition, many lung cancer cells produce growth factors that may act in an autocrine fashion on lung cancer cells. See Siegfried et al., pp. 317-336, in Kane, supra; Moody, pp. 337-370, in Kane, supra and Heasley et al., 371-390, in Kane, supra. In SCLC, many tumor cells produce gastrin-releasing peptide (GRP), which is a proliferative growth factor for these cells. See Skarin, supra. Many NSCLC tumors express epidermal growth factor (EGF) receptors, allowing NSCLC cells to proliferate in response to EGF. Insulin-like growth factor (IGF-I) is elevated in greater than 95% of SCLC and greater than 80% of NSCLC tumors; it is thought to function as an autocrine growth factor. Id. Finally, stem cell factor (SCF, also known as steel factor or kit ligand) and c-Kit (a proto-oncoprotein tyrosine kinase receptor for SCF) are both expressed at high levels in SCLC, and thus may form an autocrine loop that increases proliferation. Id.


Although the majority of lung cancer cases are attributable to cigarette smoking, most smokers do not develop lung cancer. Epidemiological evidence has suggested that susceptibility to lung cancer may be inherited in a Mendelian fashion, and thus have an inherited genetic component. Bailey-Wilson, supra. Thus, it is thought that certain allelic variants at some genetic loci may affect susceptibility to lung cancer. Id. One way to identify which allelic variants are likely to be involved in lung cancer susceptibility, as well as susceptibility to other diseases, is to look at allelic variants of genes that are highly expressed in lung.


The lung is also susceptible to a number of other debilitating diseases, including, without limitation, emphysema, pneumonia, cystic fibrosis and asthma. See Stockley (ed.), Molecular Biology of the Lung, Volume I: Emphysema and Infection, Birkhauser Verlag (1999), hereafter Stockley I, and Stockley (ed.), Molecular Biology of the Lung, Volume II: Asthma and Cancer, Birkhauser Verlag (1999), hereafter Stockley II. The cause of many these disorders is still not well understood and there are few, if any, good treatment options for many of these noncancerous lung disorders. Thus, there remains a need to understand various noncancerous lung disorders and to identify treatments for these diseases.


In yet another aspect, the development and differentiation of the lung tissue is important during embryonic development. All of the epithelial cells of the respiratory tract, including those of the lung and bronchi, are derived from the primitive endodermal cells that line the embryonic outpouching. See Yesner, supra. During embryonic development, multipotent endodermal stem cells differentiate into many different types of specialized cells, which include ciliated cells for moving inhaled particles, goblet cells for producing mucus, Kulchitsky's cells for endocrine function, and Clara cells and type II pneumocytes for secreting surfactant protein. Id. Improper development and differentiation may cause respiratory disorders and distress in infants, particularly in premature infants, whose lungs cannot produce sufficient surfactant when they are born. Further, some lung cancer cells, particularly small cell carcinomas, appear multipotent, and can spontaneously differentiate into a number of cell types, including small cell carcinoma, adenocarcinoma and squamous cell carcinoma. Id. Thus, a better understanding of lung development and differentiation may help facilitate understanding of lung cancer initiation and progression.


Accordingly, there is a great need for more sensitive and accurate methods for predicting whether a person is likely to develop lung cancer, for diagnosing lung cancer, for monitoring the progression of the disease, for staging the lung cancer, for determining whether the lung cancer has metastasized and for imaging the lung cancer. There is also a need for better treatment of lung cancer. Further, there is also a great need for diagnosing and treating noncancerous lung disorders such as emphysema, pneumonia, lung infection, pulmonary fibrosis, cystic fibrosis and asthma. There is also a need for compositions and methods of using them that can be used to identify lung tissue for forensic purposes and for determining whether a particular cell or tissue exhibits lung-specific characteristics.


In the present invention, methods are provided for detecting, diagnosing, monitoring, staging, prognosticating, imaging and treating lung cancer via lung specific genes referred to herein as LSGs. For purposes of the present invention, LSG refers, among other things, to native protein expressed by the gene comprising a polynucleotide sequence of SEQ ID NO:1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74, respectively. By “LSG” it is also meant herein polynucleotides which, due to degeneracy in genetic coding, comprise variations in nucleotide sequence as compared to SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 74 but which still encode the same polypeptide. Exemplary amino acid sequences for LSG polypeptides are set forth in SEQ ID NO: 75, 76, 77, 78, 79, 80, 81, 82, 83 and 84. In the alternative, what is meant by LSG as used herein, means the native mRNA encoded by the gene comprising the polynucleotide sequence of SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74, levels of the gene comprising the polynucleotide sequence of SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74 or levels of a polynucleotide which is capable of hybridizing under stringent conditions to the antisense sequence of SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 74.


Other objects, features, advantages and aspects of the present invention will become apparent to those of skill in the art from the following description. It should be understood, however, that the following description and the specific examples, while indicating preferred embodiments of the invention are given by way of illustration only. Various changes and modifications within the spirit and scope of the disclosed invention will become readily apparent to those skilled in the art from reading the following description and from reading the other parts of the present disclosure.


SUMMARY OF THE INVENTION

Toward these ends, and others, it is an object of the present invention to provide LSGs comprising a polynucleotide of SEQ ID NO: 1, 2,.3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74, a protein expressed by a polynucleotide of SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74, or a variant thereof which expresses the protein; or a polynucleotide which is capable of hybridizing under stringent conditions to the antisense sequence of SEQ ID NO:1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 74. Exemplary LSG polypeptides of the present invention are depicted in SEQ ID NO: 75, 76, 77, 78, 79, 80, 81, 82, 83 or 84.


It is another object of the present invention to provide a method for diagnosing the presence of lung cancer by analyzing for changes in levels of LSG in cells, tissues or bodily fluids compared with levels of LSG in preferably the same cells, tissues, or bodily fluid type of a normal human control, wherein a change in levels of LSG in the patient versus the normal human control is associated with lung cancer.


Further provided is a method of diagnosing metastatic lung cancer in a patient having lung cancer which is not known to have metastasized by identifying a human patient suspected of having lung cancer that has metastasized; analyzing a sample of cells, tissues, or bodily fluid from such patient for LSG; comparing the LSG levels in such cells, tissues, or bodily fluid with levels of LSG in preferably the same cells, tissues, or bodily fluid type of a normal human control, wherein an increase in LSG levels in the patient versus the normal human control is associated with lung cancer which has metastasized.


Also provided by the invention is a method of staging lung cancer in a human which has such cancer by identifying a human patient having such cancer; analyzing a sample of cells, tissues, or bodily fluid from such patient for LSG; comparing LSG levels in such cells, tissues, or bodily fluid with levels of LSG in preferably the same cells, tissues, or bodily fluid type of a normal human control sample, wherein an increase in LSG levels in the patient versus the normal human control is associated with a cancer which is progressing and a decrease in the levels of LSG is associated with a cancer which is regressing or in remission.


Further provided is a method of monitoring lung cancer in a human having such cancer for the onset of metastasis. The method comprises identifying a human patient having such cancer that is not known to have metastasized; periodically analyzing a sample of cells, tissues, or bodily fluid from such patient for LSG; comparing the LSG levels in such cells, tissue, or bodily fluid with levels of LSG in preferably the same cells, tissues, or bodily fluid type of a normal human control sample, wherein an increase in LSG levels in the patient versus the normal human control is associated with a cancer which has metastasized.


Further provided is a method of monitoring the change in stage of lung cancer in a human having such cancer by looking at levels of LSG in a human having such cancer. The method comprises identifying a human patient having such cancer; periodically analyzing a sample of cells, tissues, or bodily fluid from such patient for LSG; comparing the LSG levels in such cells, tissue, or bodily fluid with levels of LSG in preferably the same cells, tissues, or bodily fluid type of a normal; human control sample, wherein an increase in LSG levels in the patient versus the normal human control is associated with a cancer which is progressing and a decrease in the levels of LSG is associated with a cancer which is regressing or in remission.


Further provided are methods of designing new therapeutic agents targeted to a LSG for use in imaging and treating lung cancer. For example, in one embodiment, therapeutic agents such as antibodies targeted against LSG or fragments of such antibodies can be used to treat, detect or image localization of LSG in a patient for the purpose of detecting or diagnosing a disease or condition. In this embodiment, an increase in the amount of labeled antibody detected as compared to normal tissue would be indicative of tumor metastases or growth. Such antibodies can be polyclonal, monoclonal, or omniclonal or prepared by molecular biology techniques. The term “antibody”, as used herein and throughout the instant specification is also meant to include aptamers and single-stranded oligonucleotides such as those derived from an in vitro evolution protocol referred to as SELEX and well known to those skilled in the art. Antibodies can be labeled with a variety of detectable and therapeutic labels including, but not limited to, radioisotopes and paramagnetic metals. Therapeutic agents such as small molecules and antibodies which decrease the concentration and/or activity of LSG can also be used in the treatment of diseases characterized by overexpression of LSG. Such agents can be readily identified in accordance with teachings herein.


Other objects, features, advantages and aspects of the present invention will become apparent to those of skill in the art from the following description. It should be understood, however, that the following description and the specific examples, while indicating preferred embodiments of the invention, are given by way of illustration only. Various changes and modifications within the spirit and scope of the disclosed invention will become readily apparent to those skilled in the art from reading the following description and from reading the other parts of the present disclosure.


Glossary


The following illustrative explanations are provided to facilitate understanding of certain terms used frequently herein, particularly in the examples. The explanations are provided as a convenience and are not limitative of the invention.


ISOLATED means altered “by the hand of man” from its natural state; i.e., that, if it occurs in nature, it has been changed or removed from its original environment, or both.


For example, a naturally occurring polynucleotide or a polypeptide naturally present in a living animal in its natural state is not “isolated,” but the same polynucleotide or polypeptide separated from the coexisting materials of its natural state is “isolated”, as the term is employed herein. For example, with respect to polynucleotides, the term isolated means that it is separated from the chromosome and cell in which it naturally occurs.


As part of or following isolation, such polynucleotides can be joined to other polynucleotides, such as DNAS, for mutagenesis, to form fusion proteins, and for propagation or expression in a host, for instance. The isolated polynucleotides, alone or joined to other polynucleotides such as vectors, can be introduced into host cells, in culture or in whole organisms. When introduced into host cells in culture or in whole organisms, such DNAs still would be isolated, as the term is used herein, because they would not be in their naturally occurring form or environment. Similarly, the polynucleotides and polypeptides may occur in a composition, such as media formulations, solutions for introduction of polynucleotides or polypeptides, for example, into cells, compositions or solutions for chemical or enzymatic reactions, for instance, which are not naturally occurring compositions, and, therein remain isolated polynucleotides or polypeptides within the meaning of that term as it is employed herein.


OLIGONUCLEOTIDE(S) refers to relatively short polynucleotides. Often the term refers to single-stranded deoxyribonucleotides, but it can refer as well to single-or double-stranded ribonucleotides, RNA:DNA hybrids and double-stranded DNAs, among others.


Oligonucleotides, such as single-stranded DNA probe oligonucleotides, often are synthesized by chemical methods, such as those implemented on automated oligonucleotide synthesizers. However, oligonucleotides can be made by a variety of other methods, including in vitro recombinant DNA-mediated techniques and by expression of DNAs in cells and organisms.


Initially, chemically synthesized DNAs typically are obtained without a 5′ phosphate. The 5′ ends of such oligonucleotides are not substrates for phosphodiester bond formation by ligation reactions that employ DNA ligases typically used to form recombinant DNA molecules. Where ligation of such oligonucleotides is desired, a phosphate can be added by standard techniques, such as those that employ a kinase and ATP.


The 3′ end of a chemically synthesized oligonucleotide generally has a free hydroxyl group and, in the presence of a ligase such as T4 DNA ligase, readily will form a phosphodiester bond with a 5′ phosphate of another polynucleotide, such as another oligonucleotide. As is well known, this reaction can be prevented selectively, where desired, by removing the 5′ phosphates of the other polynucleotide(s) prior to ligation.


POLYNUCLEOTIDE(S) generally refers to any polyribonucleotide or polydeoxribonucleotide and is inclusive of unmodified RNA or DNA as well as modified RNA or DNA. Thus, for instance, polynucleotides as used herein refers to, among other things, single- and double-stranded DNA, DNA that is a mixture of single- and double-stranded regions, single- and double-stranded RNA, and RNA that is mixture of single- and double-stranded regions, hybrid molecules comprising DNA and RNA that may be single-stranded or, more typically, double-stranded or a mixture of single- and double-stranded regions. In addition, polynucleotide, as used herein, refers to triple-stranded regions comprising RNA or DNA or both RNA and DNA. The strands in such regions may be from the same molecule or from different molecules. The regions may include all of one or more of the molecules, but more typically involve only a region of some of the molecules. One of the molecules of a triple-helical region often is an oligonucleotide.


As used herein, the term polynucleotide is also inclusive of DNAs or RNAs as described above that contain one or more modified bases. Thus, DNAs or RNAs with backbones modified for stability or for other reasons are “polynucleotides” as that term is intended herein. Moreover, DNAs or RNAs comprising unusual bases, such as inosine, or modified bases, such as tritylated bases, to name just two examples, are polynucleotides as the term is used herein.


It will be appreciated that a great variety of modifications have been made to DNA and RNA that serve many useful purposes known to those of skill in the art. The term polynucleotide as it is employed herein embraces such chemically, enzymatically or metabolically modified forms of polynucleotides, as well as chemical forms of DNA and RNA characteristic of viruses and cells, including simple and complex cells, inter alia.


POLYPEPTIDES, as used herein, includes all polypeptides as described below. The basic structure of polypeptides is well known and has been described in innumerable textbooks and other publications in the art. In this context, the term is used herein to refer to any peptide or protein comprising two or more amino acids joined to each other in a linear chain by peptide bonds. As used herein, the term refers to both short chains, which also commonly are referred to in the art as peptides, oligopeptides and oligomers, for example, and to longer chains, which generally are referred to in the art as proteins, of which there are many types. It will be appreciated that polypeptides often contain amino acids other than the 20 amino acids commonly referred to as the naturally occurring amino acids, and that many amino acids, including the terminal amino acids, may be modified in a given polypeptide, either by natural processes such as processing and other post-translational modifications, or by chemical modification techniques which are well known to the art. Even the common modifications that occur naturally in polypeptides are too numerous to list exhaustively here, but they are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature, and they are well known to those of skill in the art.


Modifications which may be present in polypeptides of the present invention include, to name an illustrative few, acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cystine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination.


Such modifications are well known to those of skill and have been described in great detail in the scientific literature. Several particularly common modifications including, but not limited to, glycosylation, lipid attachment, sulfation, gamma-carboxylation of glutamic acid residues, hydroxylation and ADP-ribosylation are described in most basic texts, such as, for instance PROTEINS STRUCTURE AND MOLECULAR PROPERTIES, 2nd Ed., T. E. Creighton, W. H. Freeman and Company, New York (1993). Many detailed reviews are available on this subject, such as, for example, those provided by Wold, F., Posttranslational Protein Modifications: Perspectives and Prospects, pgs. 1-12 in POSTTRANSLATIONAL COVALENT MODIFICATION OF PROTEINS, B. C. Johnson, Ed., Academic Press, New York (1983); Seifter et al., Analysis for protein modifications and nonprotein cofactors, Meth. Enzymol. 182: 626-646 (1990) and Rattan et al., Protein Synthesis: Posttranslational Modifications and Aging, Ann. N.Y. Acad. Sci. 663: 48-62 (1992).


It will be appreciated that the polypeptides of the present invention are not always entirely linear. Instead, polypeptides may be branched as a result of ubiquitination, and they may be circular, with or without branching, generally as a result of posttranslation events including natural processing event and events brought about by human manipulation which do not occur naturally. Circular, branched and branched circular polypeptides may be synthesized by non-translation natural processes and by entirely synthetic methods, as well.


Modifications can occur anywhere in a polypeptide, including the peptide backbone, the amino acid side-chains and the amino or carboxyl termini. In fact, blockage of the amino and/or carboxyl group in a polypeptide by a covalent modification is common in naturally occurring and synthetic polypeptides and such modifications may be present in polypeptides of the present invention, as well. For instance, the amino terminal residue of polypeptides made in E. coli, prior to proteolytic processing, almost invariably will be N-formylmethionine.


The modifications that occur in a polypeptide often will be a function of how it is made. For polypeptides made by expressing a cloned gene in a host, for instance, the nature and extent of the modifications, in large part, will be determined by the host cell posttranslational modification capacity and the modification signals present in the polypeptide amino acid sequence. For instance, as is well known, glycosylation often does not occur in bacterial hosts such as E. coli. Accordingly, when glycosylation is desired, a polypeptide can be expressed in a glycosylating host, generally a eukaryotic cell. Insect cells often carry out the same posttranslational glycosylations as mammalian cells. Thus, insect cell expression systems have been developed to express efficiently mammalian proteins having native patterns of glycosylation, inter alia. Similar considerations apply to other modifications.


It will be appreciated that the same type of modification may be present in the same or varying degrees at several sites in a given polypeptide. Also, a given polypeptide may contain many types of modifications.


In general, as used herein, the term polypeptide encompasses all such modifications, particularly those that are present in polypeptides synthesized by expressing a polynucleotide in a host cell.


VARIANT(S) of polynucleotides or polypeptides, as the term is used herein, are polynucleotides or polypeptides that differ from a reference polynucleotide or polypeptide, respectively.


With respect to variant polynucleotides, differences are generally limited so that the nucleotide sequences of the reference and the variant are closely similar overall and, in many regions, identical. Thus, changes in the nucleotide sequence of the variant may be silent. That is, they may not alter the amino acids encoded by the polynucleotide. Where alterations are limited to silent changes of this type a variant will encode a polypeptide with the same amino acid sequence as the reference. Alternatively, changes in the nucleotide sequence of the variant may alter the amino acid sequence of a polypeptide encoded by the reference polynucleotide. Such nucleotide changes may result in amino acid substitutions, additions, deletions, fusions and truncations in the polypeptide encoded by the reference sequence.


With respect to variant polypeptides, differences are generally limited so that the sequences of the reference and the variant are closely similar overall and, in many region, identical. For example, a variant and reference polypeptide may differ in amino acid sequence by one or more substitutions, additions, deletions, fusions and truncations, which may be present in any combination.


RECEPTOR MOLECULE, as used herein, refers to molecules which bind or interact specifically with LSG polypeptides of the present invention and is inclusive not only of classic receptors, which are preferred, but also other molecules that specifically bind to or interact with polypeptides of the invention (which also may be referred to as “binding molecules” and “interaction molecules,” respectively and as “LSG binding or interaction molecules”. Binding between polypeptides of the invention and such molecules, including receptor or binding or interaction molecules may be exclusive to polypeptides of the invention, which is very highly preferred, or it may be highly specific for polypeptides of the invention, which is highly preferred, or it may be highly specific to a group of proteins that includes polypeptides of the invention, which is preferred, or it may be specific to several groups of proteins at least one of which includes polypeptides of the invention.


Receptors also may be non-naturally occurring, such as antibodies and antibody-derived reagents that bind to polypeptides of the invention.







DETAILED DESCRIPTION OF THE INVENTION

The present invention relates to novel lung specific polypeptides and polynucleotides, referred to herein as LSGs, among other things, as described in greater detail below.


Polynucleotides


In accordance with one aspect of the present invention, there are provided isolated LSG polynucleotides which encode LSG polypeptides.


Using the information provided herein, such as the polynucleotide sequences set out in SEQ ID NO:1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 74 a polynucleotide of the present invention encoding a LSG may be obtained using standard cloning and screening procedures, such as those for cloning cDNAs using mRNA from cells of a human tumor as starting material.


Polynucleotides of the present invention may be in the form of RNA, such as mRNA, or in the form of DNA, including, for instance, cDNA and genomic DNA obtained by cloning or produced by chemical synthetic techniques or by a combination thereof. The DNA may be double-stranded or single-stranded. Single-stranded DNA may be the coding strand, also known as the sense strand, or it may be the non-coding strand, also referred to as the anti-sense strand.


The coding sequence which encodes the polypeptides may be identical to the coding sequence of the polynucleotides of SEQ ID NO:1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74. It also may be a polynucleotide with a different sequence, which, as a result of the redundancy (degeneracy) of the genetic code, encodes the same polypeptides as encoded by SEQ ID NO:1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74.


Polynucleotides of the present invention, such as SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74 which encode these polypeptides may comprise the coding sequence for the mature polypeptide by itself. Polynucleotides of the present invention may also comprise the coding sequence for the mature polypeptide and additional coding sequences such as those encoding a leader or secretory sequence such as a pre-, or pro- or prepro-protein sequence. Polynucleotides of the present invention may also comprise the coding sequence of the mature polypeptide, with or without the aforementioned additional coding sequences, together with additional, non-coding sequences. Examples of additional non-coding sequences which may be incorporated into the polynucleotide of the present invention include, but are not limited to, introns and non-coding 5′ and 3′ sequences such as transcribed, non-translated sequences that play a role in transcription, mRNA processing including, for example, splicing and polyadenylation signals, ribosome binding and stability of mRNA, and additional coding sequence which codes for amino acids such as those which provide additional functionalities. Thus, for instance, the polypeptide may be fused to a marker sequence such as a peptide which facilitates purification of the fused polypeptide. In certain preferred embodiments of this aspect of the invention, the marker sequence is a hexa-histidine peptide, such as the tag provided in the pQE vector (Qiagen, Inc.), among others, many of which are commercially available. As described in Gentz et al. (Proc. Natl. Acad. Sci., USA 86: 821-824 (1989)), for instance, hexa-histidine provides for convenient purification of the fusion protein. The HA tag corresponds to an epitope derived of influenza hemagglutinin protein (Wilson et al., Cell 37: 767 (1984)).


In accordance with the foregoing, the term “polynucleotide encoding a polypeptide” as used herein encompasses polynucleotides which include a sequence encoding a polypeptide of the present invention, particularly SEQ ID NO:1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74. Exemplary polypeptides encoded by the polynucleotides are depicted in SEQ ID NO: 75, 76, 77, 78, 79, 80, 81, 82, 83 and 84. The term encompasses polynucleotides that include a single continuous region or discontinuous regions encoding the polypeptide (for example, interrupted by introns) together with additional regions, that also may contain coding and/or non-coding sequences.


The present invention further relates to variants of the herein above described polynucleotides which encode for fragments, analogs and derivatives of the LSG polypeptides. A variant of the polynucleotide may be a naturally occurring variant such as a naturally occurring allelic variant, or it may be a variant that is not known to occur naturally. Such non-naturally occurring variants of the polynucleotide may be made by mutagenesis techniques, including those applied to polynucleotides, cells or organisms.


Among variants in this regard are variants that differ from the aforementioned polynucleotides by nucleotide substitutions, deletions or additions. The substitutions, deletions or additions may involve one or more nucleotides. The variants may be altered in coding or non-coding regions or both. Alterations in the coding regions may produce conservative or non-conservative amino acid substitutions, deletions or additions.


Among the particularly preferred embodiments of the invention in this regard are polynucleotides encoding polypeptides having the same amino acid sequence encoded by a LSG polynucleotide comprising SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74; variants, analogs, derivatives and fragments thereof, and fragments of the variants, analogs and derivatives. Exemplary polypeptides encoded by these polynucleotides are depicted in SEQ ID NO:75, 76, 77, 78, 79, 80, 81, 82, 83 and 84. Further particularly preferred in this regard are LSG polynucleotides encoding polypeptide variants, analogs, derivatives and fragments, and variants, analogs and derivatives of the fragments, in which several, a few, 5 to 10, 1 to 5, 1 to 3, 2, 1 or no amino acid residues are substituted, deleted or added, in any combination. Especially preferred among these are silent substitutions, additions and deletions, which do not alter the properties and activities of the LSG. Also especially preferred in this regard are conservative substitutions. Most highly preferred are polynucleotides encoding polypeptides having the amino acid sequences as polypeptides encoded by SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74, without substitutions.


Further preferred embodiments of the invention are LSG polynucleotides that are at least 70% identical to a polynucleotide of SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74 and polynucleotides which are complementary to such polynucleotides. More preferred are LSG polynucleotides that comprise a region that is at least 80% identical to a polynucleotide of SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74. In this regard, LSG polynucleotides at least 90% identical to the same are particularly preferred, and among these particularly preferred LSG polynucleotides, those with at least 95% are especially preferred. Furthermore, those with at least 97% are highly preferred among those with at least 95%, and among these those with at least 98% and at least 99% are particularly highly preferred, with at least 99% being the most preferred.


Particularly preferred embodiments in this respect, moreover, are polynucleotides which encode polypeptides which retain substantially the same biological function or activity as the mature polypeptides encoded by a polynucleotide of SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74.


The present invention further relates to polynucleotides that hybridize to the herein above-described LSG sequences. In this regard, the present invention especially relates to polynucleotides which hybridize under stringent conditions to the herein above-described polynucleotides. As herein used, the term “stringent conditions” means hybridization will occur only if there is at least 95% and preferably at least 97% identity between the sequences.


As discussed additionally herein regarding polynucleotide assays of the invention, for instance, polynucleotides of the invention as described herein, may be used as a hybridization probe for cDNA and genomic DNA to isolate full-length cDNAs and genomic clones encoding


LSGs and to isolate cDNA and genomic clones of other genes that have a high sequence similarity to these LSGs. Such probes generally will comprise at least 15 bases. Preferably, such probes will have at least 30 bases and may have at least 50 bases.


For example, the coding region of LSG of the present invention may be isolated by screening using an oligonucleotide probe synthesized from the known DNA sequence. A labeled oligonucleotide having a sequence complementary to that of a gene of the present invention is used to screen a library of human cDNA, genomic DNA or mRNA to determine which members of the library the probe hybridizes with.


The polynucleotides and polypeptides of the present invention may be employed as research reagents and materials for discovery of treatments and diagnostics to human disease, as further discussed herein relating to polynucleotide assays, inter alia.


The polynucleotides may encode a polypeptide which is the mature protein plus additional amino or carboxyl-terminal amino acids, or amino acids interior to the mature polypeptide (when the mature form has more than one polypeptide chain, for instance). Such sequences may play a role in processing of a protein from precursor to a mature form, may facilitate/protein trafficking, may prolong or shorten protein half-life or may facilitate manipulation of a protein for assay or production, among other things. As generally is the case in situ, the additional amino acids may be processed away from the mature protein by cellular enzymes.


A precursor protein having the mature form of the polypeptide fused to one or more prosequences may be an inactive form of the polypeptide. When prosequences are removed, such inactive precursors generally are activated. Some or all of the prosequences may be removed before activation. Generally, such precursors are called proproteins.


In sum, a polynucleotide of the present invention may encode a mature protein, a mature protein plus a leader sequence (which may be referred to as a preprotein), a precursor of a mature protein having one or more prosequences which are not the leader sequences of a preprotein, or a preproprotein, which is a precursor to a proprotein, having a leader sequence and one or more prosequences, which generally are removed during processing steps that produce active and mature forms of the polypeptide.


Polypeptides


The present invention further relates to LSG polypeptides, preferably polypeptides encoded by a polynucleotide of SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74. Exemplary polypeptides are depicted in SEQ ID NO: 75, 76, 77, 78, 79, 80, 81, 82, 83 or 84. The invention also relates to fragments, analogs and derivatives of these polypeptides. The terms “fragment,” “derivative” and “analog” when referring to the polypeptides of the present invention means a polypeptide which retains essentially the same biological function or activity as such-polypeptides. Thus, an analog includes a proprotein which can be activated by cleavage of the proprotein portion to produce an active mature polypeptide.


The polypeptide of the present invention may be a recombinant polypeptide, a natural polypeptide or a synthetic polypeptide. In certain preferred embodiments it is a recombinant polypeptide.


The fragment, derivative or analog of a polypeptide of or the present invention may be (I) one in which one or more of the amino acid residues are substituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue) and such substituted amino acid residue may or may not be one encoded by the genetic code; (ii) one in which one or more of the amino acid residues includes a substituent group; (iii) one in which the mature polypeptide is fused with another compound, such as a compound to increase the half-life of the polypeptide (for example, polyethylene glycol); or (iv) one in which the additional amino acids are fused to the mature polypeptide, such as a leader or secretory sequence or a sequence which is employed for purification of the mature polypeptide or a proprotein sequence. Such fragments, derivatives and analogs are deemed to be within the scope of those skilled in the art from the teachings herein.


Among preferred variants are those that vary from a reference by conservative amino acid substitutions. Such substitutions are those that substitute a given amino acid in a polypeptide by another amino acid of like characteristics. Typically seen as conservative substitutions are the replacements, one for another, among the aliphatic amino acids Ala, Val, Leu and Ile; interchange of the hydroxyl residues Ser and Thr, exchange of the acidic residues Asp and Glu, substitution between the amide residues Asn and Gln, exchange of the basic residues Lys and Arg and replacements among the aromatic residues Phe, Tyr.


The polypeptides and polynucleotides of the present invention are preferably provided in an isolated form, and preferably are purified to homogeneity.


The polypeptides of the present invention include the polypeptides encoded by the polynucleotide of SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74 (in particular the mature polypeptide) as well as polypeptides which have at least 75% similarity (preferably at least 75% identity), more preferably at least 90% similarity (more preferably at least 90% identity), still more preferably at least 95% similarity (still more preferably at least 95% identity), to a polypeptide encoded by SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74. Also included are portions of such polypeptides generally containing at least 30 amino acids and more preferably at least 50 amino acids. Exemplary polypeptides are depicted in SEQ ID NO:75, 76, 77, 78, 79, 80, 81, 82, 83 or 84.


As known in the art “similarity” between two polypeptides is determined by comparing the amino acid sequence and its conserved amino acid substitutes of one polypeptide sequence with that of a second polypeptide.


Fragments or portions of the polypeptides of the present invention may be employed for producing the corresponding full-length polypeptide by peptide synthesis; therefore, the fragments may be employed as intermediates for producing the full-length polypeptides. Fragments or portions of the polynucleotides of the present invention may be used to synthesize full-length polynucleotides of the present invention.


Fragments


Also among preferred embodiments of this aspect of the present invention are polypeptides comprising fragments of a polypeptide encoded by a polynucleotide of SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74. In this regard a fragment is a polypeptide having an amino acid sequence that entirely is the same as part but not all of the amino acid sequence of the aforementioned LSG polypeptides and variants or derivatives thereof.


Such fragments may be “free-standing,” i.e., not part of or fused to other amino acids or polypeptides, or they may be contained within a larger polypeptide of which they form a part or region. When contained within a larger polypeptide, the presently discussed fragments most preferably form a single continuous region. However, several fragments may be comprised within a single larger polypeptide. For instance, certain preferred embodiments relate to a fragment of a LSG polypeptide of the present comprised within a precursor polypeptide designed for expression in a host and having heterologous pre- and pro-polypeptide regions fused to the amino terminus of the LSG fragment and an additional region fused to the carboxyl terminus of the fragment. Therefore, fragments in one aspect of the meaning intended herein, refers to the portion or portions of a fusion polypeptide or fusion protein derived from a LSG polypeptide.


As representative examples of polypeptide fragments of the invention, there may be mentioned those which have from about 15 to about 139 amino acids. In this context “about” includes the particularly recited range and ranges larger or smaller by several, a few, 5, 4, 3, 2 or 1 amino acid at either extreme or at both extremes. Highly preferred in this regard are the recited ranges plus or minus as many as 5 amino acids at either or at both extremes. Particularly highly preferred are the recited ranges plus or minus as many as 3 amino acids at either or at both the recited extremes. Especially preferred are ranges plus or minus 1 amino acid at either or at both extremes or the recited ranges with no additions or deletions. Most highly preferred of all in this regard are fragments from about 15 to about 45 amino acids.


Among especially preferred fragments of the invention are truncation mutants of the LSG polypeptides. Truncation mutants include LSG polypeptides having an amino acid sequence encoded by a polynucleotide of SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74 or variants or derivatives thereof, except for deletion of a continuous series of residues (that is, a continuous region, part or portion) that includes the amino terminus, or a continuous series of residues that includes the carboxyl terminus or, as in double truncation mutants, deletion of two continuous series of residues, one including the amino terminus and one including the carboxyl terminus. Fragments having the size ranges set out herein also are preferred embodiments of truncation fragments, which are especially preferred among fragments generally.


Also preferred in this aspect of the invention are fragments characterized by structural or functional attributes of the LSG polypeptides of the present invention. Preferred embodiments of the invention in this regard include fragments that comprise alpha-helix and alpha-helix forming regions (“alpha-regions”), beta-sheet and beta-sheet-forming regions (“beta-regions”), turn and turn-forming regions (“turn-regions”), coil and coil-forming regions (“coil-regions”), hydrophilic regions, hydrophobic regions, alpha amphipathic regions, beta amphipathic regions, flexible regions, surface-forming regions and high antigenic index regions of the LSG polypeptides of the present invention. Regions of the aforementioned types are identified routinely by analysis of the amino acid sequences encoded by the polynucleotides of SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74. Preferred regions include Garnier-Robson alpha-regions, beta-regions, turn-regions and coil-regions, Chou-Fasman alpha-regions, beta-regions and turn-regions, Kyte-Doolittle hydrophilic regions and hydrophilic regions, Eisenberg alpha and beta amphipathic regions, Karplus-Schulz flexible regions, Emini surface-forming regions and Jameson-Wolf high antigenic index regions. Among highly preferred fragments in this regard are those that comprise regions of LSGs that combine several structural features, such as several of the features set out above. In this regard, the regions defined by selected residues of a LSG polypeptide which all are characterized by amino acid compositions highly characteristic of turn-regions, hydrophilic regions, flexible-regions, surface-forming regions, and high antigenic index-regions, are especially highly preferred regions. Such regions may be comprised within a larger polypeptide or may be by themselves a preferred fragment of the present invention, as discussed above. It will be appreciated that the term “about” as used in this paragraph has the meaning set out above regarding fragments in general.


Further preferred regions are those that mediate activities of LSG polypeptides. Most highly preferred in this regard are fragments that have a chemical, biological or other activity of a LSG polypeptide, including those with a similar activity or an improved activity, or with a decreased undesirable activity. Highly preferred in this regard are fragments that contain regions that are homologs in sequence, or in position, or in both sequence and to active regions of related polypeptides, and which include lung specific-binding proteins. Among particularly preferred fragments in these regards are truncation mutants, as discussed above.


It will be appreciated that the invention also relates to polynucleotides encoding the aforementioned fragments, polynucleotides that hybridize to polynucleotides encoding the fragments, particularly those that hybridize under stringent conditions, and polynucleotides such as PCR primers for amplifying polynucleotides that encode the fragments. In these regards, preferred polynucleotides are those that correspond to the preferred fragments, as discussed above.


Fusion Proteins


In one embodiment of the present invention, the LSG polypeptides of the present invention are preferably fused to other proteins. These fusion proteins can be used for a variety of applications. For example, fusion of the present polypeptides to His-tag, HA-tag, protein A, IgG domains, and maltose binding protein facilitates purification. (See also EP A 394,827; Traunecker, et al., Nature 331: 84-86 (1988)) Similarly, fusion to IgG-1, IgG-3, and albumin increases the halflife time in vivo. Nuclear localization signals fused to the polypeptides of the present invention can target the protein to a specific subcellular localization, while covalent heterodimer or homodimers can increase or decrease the activity of a fusion protein. Fusion proteins can also create chimeric molecules having more than one function. Finally, fusion proteins can increase solubility and/or stability of the fused protein compared to the non-fused protein. All of these types of fusion proteins described above can be made in accordance with well known protocols.


For example, a LSG polypeptide can be fused to an IgG molecule via the following protocol. Briefly, the human Fc portion of the IgG molecule is PCR amplified using primers that span the 5′ and 3′ ends of the sequence. These primers also have convenient restriction enzyme sites that facilitate cloning into an expression vector, preferably a mammalian expression vector. For example, if pC4 (Accession No. 209646) is used, the human Fc portion can be ligated into the BamHI cloning site. In this protocol, the 3′ BamHI site must be destroyed. Next, the vector containing the human Fc portion is re-restricted with BamHI thereby linearizing the vector, and a LSG polynucleotide of the present invention is ligated into this BamHI site. It is preferred that the polynucleotide is cloned without a stop codon, otherwise a fusion protein will not be produced.


If the naturally occurring signal sequence is used to produce the secreted protein, pC4 does not need a second signal peptide. Alternatively, if the naturally occurring signal sequence is not used, the vector can be modified to include a heterologous signal sequence. (See, e. g., WO 96/34891.)


Diagnostic Assays


The present invention also relates to diagnostic assays and methods, both quantitative and qualitative for detecting, diagnosing, monitoring, staging and prognosticating cancers by comparing levels of LSG in a human patient with those of LSG in a normal human control. For purposes of the present invention, what is meant by LSG levels is, among other things, native protein expressed by a gene comprising the polynucleotide sequence of SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74. Exemplary polypeptides encoded by these polynucleotides are depicted in SEQ ID NO:75, 76, 77, 78, 79, 80, 81, 82, 83 and 84. By “LSG” it is also meant herein polynucleotides which, due to degeneracy in genetic coding, comprise variations in nucleotide sequence as compared to SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74 but which still encode the same protein. The native protein being detected may be whole, a breakdown product, a complex of molecules or chemically modified. In the alternative, what is meant by LSG as used herein, means the native mRNA encoded by a polynucleotide sequence of SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74, levels of the gene comprising the polynucleotide sequence of SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74, or levels of a polynucleotide which is capable of hybridizing under stringent conditions to the antisense sequence of SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74. Such levels are preferably determined in at least one of cells, tissues and/or bodily fluids, including determination of normal and abnormal levels. Thus, for instance, a diagnostic assay in accordance with the invention for diagnosing overexpression of LSG protein compared to normal control bodily fluids, cells, or tissue samples may be used to diagnose the presence of lung cancer.


All the methods of the present invention may optionally include determining the levels of other cancer markers as well as LSG. Other cancer markers, in addition to LSG, useful in the present invention will depend on the cancer being tested and are known to those of skill in the art.


The present invention provides methods for diagnosing the presence of lung cancer by analyzing for changes in levels of LSG in cells, tissues or bodily fluids compared with levels of LSG in cells, tissues or bodily fluids of preferably the same type from a normal human control, wherein an increase in levels of LSG in the patient versus the normal human control is associated with the presence of lung cancer.


Without limiting the instant invention, typically, for a quantitative diagnostic assay a positive result indicating the patient being tested has cancer is one in which cells, tissues or bodily fluid levels of the cancer marker, such as LSG, are at least two times higher, and most preferably are at least five times higher, than in preferably the same cells, tissues or bodily fluid of a normal human control.


The present invention also provides a method of diagnosing metastatic lung cancer in a patient having lung cancer which has not yet metastasized for the onset of metastasis. In the method of the present invention, a human cancer patient suspected of having lung cancer which may have metastasized (but which was not previously known to have metastasized) is identified. This is accomplished by a variety of means known to those of skill in the art.


In the present invention, determining the presence of LSG levels in cells, tissues or bodily fluid, is particularly useful for discriminating between lung cancer which has not metastasized and lung cancer which has metastasized. Existing techniques have difficulty discriminating between lung cancer which has metastasized and lung cancer which has not metastasized and proper treatment selection is often dependent upon such knowledge.


In the present invention, the cancer marker levels measured in such cells, tissues or bodily fluid is LSG, and are compared with levels of LSG in preferably the same cells, tissue or bodily fluid type of a normal human control. That is, if the cancer marker being observed is just LSG in serum, this level is preferably compared with the level of LSG in serum of a normal human control. An increase in the LSG in the patient versus the normal human control is associated with lung cancer which has metastasized.


Without limiting the instant invention, typically, for a quantitative diagnostic assay a positive result indicating the cancer in the patient being tested or monitored has metastasized is one in which cells, tissues or bodily fluid levels of the cancer marker, such as LSG, are at least two times higher, and most preferably are at least five times higher, than in preferably the same cells, tissues or bodily fluid of a normal patient.


Normal human control as used herein includes a human patient without cancer and/or non cancerous samples from the patient; in the methods for diagnosing or monitoring for metastasis, normal human control may preferably also include samples from a human patient that is determined by reliable methods to have lung cancer which has not metastasized.


Staging


The invention also provides a method of staging lung cancer in a human patient. The method comprises identifying a human patient having such cancer and analyzing cells, tissues or bodily fluid from such human patient for LSG. The LSG levels determined in the patient are then compared with levels of LSG in preferably the same cells, tissues or bodily fluid type of a normal human control, wherein an increase in LSG levels in the human patient versus the normal human control is associated with a cancer which is progressing and a decrease in the levels of LSG (but still increased over true normal levels) is associated with a cancer which is regressing or in remission.


Monitoring


Further provided is a method of monitoring lung cancer in a human patient having such cancer for the onset of metastasis. The method comprises identifying a human patient having such cancer that is not known to have metastasized; periodically analyzing cells, tissues or bodily fluid from such human patient for LSG; and comparing the LSG levels determined in the human patient with levels of LSG in preferably the same cells, tissues or bodily fluid type of a normal human control, wherein an increase in LSG levels in the human patient versus the normal human control is associated with a cancer which has metastasized. In this method, normal human control samples may also include prior patient samples.


Further provided by this invention is a method of monitoring the change in stage of lung cancer in a human patient having such cancer. The method comprises identifying a human patient having such cancer; periodically analyzing cells, tissues or bodily fluid from such human patient for LSG; and comparing the LSG levels determined in the human patient with levels of LSG in preferably the same cells, tissues or bodily fluid type of a normal human control, wherein an increase in LSG levels in the human patient versus the normal human control is associated with a cancer which is progressing in stage and a decrease in the levels of LSG is associated with a cancer which is regressing in stage or in remission. In this method, normal human control samples may also include prior patient samples.


Monitoring a patient for onset of metastasis is periodic and preferably done on a quarterly basis.


However, this may be done more or less frequently depending on the cancer, the particular patient, and the stage of the cancer.


Prognostic Testing and Clinical Trial Monitoring


The methods described herein can further be utilized as prognostic assays to identify subjects having or at risk of developing a disease or disorder associated with increased levels of LSG. The present invention provides a method in which a test sample is obtained from a human patient and LSG is detected. The presence of higher LSG levels as compared to normal human controls is diagnostic for the human patient being at risk for developing cancer, particularly lung cancer.


The effectiveness of therapeutic agents to decrease expression or activity of the LSGs of the invention can also be monitored by analyzing levels of expression of the LSGs in a human patient in clinical trials or in in vitro screening assays such as in human cells. In this way, the gene expression pattern can serve as a marker, indicative of the physiological response of the human patient, or cells as the case may be, to the agent being tested.


Detection of Genetic Lesions or Mutations


The methods of the present invention can also be used to detect genetic lesions or mutations in LSG, thereby determining if a human with the genetic lesion is at risk for lung cancer or has lung cancer. Genetic lesions can be detected, for example, by ascertaining the existence of a deletion and/or addition and/or substitution of one or more nucleotides from the LSGs of this invention, a chromosomal rearrangement of LSG, aberrant modification of LSG (such as of the methylation pattern of the genomic DNA), the presence of a non-wild type splicing pattern of a mRNA transcript of LSG, allelic loss of LSG, and/or inappropriate post-translational modification of LSG protein. Methods to detect such lesions in the LSG of this invention are known to those of skill in the art.


For example, in one embodiment, alterations in a gene corresponding to a LSG polynucleotide of the present invention are determined via isolation of RNA from entire families or individual patients presenting with a phenotype of interest (such as a disease) is be isolated. cDNA is then generated from these RNA samples using protocols known in the art. See, e.g. Sambrook et al. (MOLECULAR CLONING: A LABORATORY MANUAL, 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)), which is illustrative of the many laboratory manuals that detail these techniques. The cDNA is then used as a template for PCR, employing primers surrounding regions of interest in SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74. PCR conditions typically consist of 35 cycles at 95° C. for 30 seconds; 60-120 seconds at 52-58° C.; and 60-120 seconds at 70° C., using buffer solutions described in Sidransky, D., et al., Science 252: 706 (1991). PCR products are sequenced using primers labeled at their 5′ end with T4 polynucleotide kinase, employing SequiTherm Polymerase (Epicentre Technologies). The intron-exon borders of selected exons are also determined and genomic PCR products analyzed to confirm the results. PCR products harboring suspected mutations are then cloned and sequenced to validate the results of the direct sequencing. PCR products are cloned into T-tailed vectors as described in Holton, T. A. and Graham, M. W., Nucleic Acids Research, 19:1156 (1991) and sequenced with T7 polymerase (United States Biochemical). Affected individuals are identified by mutations not present in unaffected individuals.


Genomic rearrangements can also be observed as a method of determining alterations in a gene corresponding to a polynucleotide. In this method, genomic clones are nick-translated with digoxigenin deoxy-uridine 5′ triphosphate (Boehringer Manheim), and FISH is performed as described in Johnson, C. et al., Methods Cell Biol. 35: 73-99 (1991). Hybridization with a labeled probe is carried out using a vast excess of human DNA for specific hybridization to the corresponding genomic locus. Chromosomes are counterstained with 4,6-diamino-2-phenylidole and propidium iodide, producing a combination of C— and R-bands. Aligned images for precise mapping are obtained using a triple-band filter set (Chroma Technology, Brattleboro, Vt.) in combination with a cooled charge-coupled device camera (Photometrics, Tucson, Ariz.) and variable excitation wavelength filters (Johnson et al., Genet. Anal. Tech. Appl., 8: 75 (1991)). Image collection, analysis and chromosomal fractional length measurements are performed using the ISee Graphical Program System (Inovision Corporation, Durham, N.C.). Chromosome alterations of the genomic region hybridized by the probe are identified as insertions, deletions, and translocations. These alterations are used as a diagnostic marker for an associated disease.


Assay Techniques


Assay techniques that can be used to determine levels of gene expression (including protein levels), such as LSG of the present invention, in a sample derived from a patient are well known to those of skill in the art. Such assay methods include, without limitation, radioimmunoassays, reverse transcriptase PCR (RT-PCR) assays, immunohistochemistry assays, in situ hybridization assays, competitive-binding assays, Western Blot analyses, ELISA assays and proteomic approaches: two-dimensional gel electrophoresis (2D electrophoresis) and non-gel based approaches such as mass spectrometry or protein interaction profiling. Among these, ELISAs are frequently preferred to diagnose a gene's expressed protein in biological fluids.


An ELISA assay initially comprises preparing an antibody, if not readily available from a commercial source, specific to LSG, preferably a monoclonal antibody. In addition a reporter antibody generally is prepared which binds specifically to LSG. The reporter antibody is attached to a detectable reagent such as radioactive, fluorescent or enzymatic reagent, for example horseradish peroxidase enzyme or alkaline phosphatase.


To carry out the ELISA, antibody specific to LSG is incubated on a solid support, e.g. a polystyrene dish, that binds the antibody. Any free protein binding sites on the dish are then covered by incubating with a non-specific protein such as bovine serum albumin. Next, the sample to be analyzed is incubated in the dish, during which time LSG binds to the specific antibody attached to the polystyrene dish. Unbound sample is washed out with buffer. A reporter antibody specifically directed to LSG and linked to a detectable reagent such as horseradish peroxidase is placed in the dish resulting in binding of the reporter antibody to any monoclonal antibody bound to LSG. Unattached reporter antibody is then washed out. Reagents for peroxidase activity, including a calorimetric substrate are then added to the dish. Immobilized peroxidase, linked to LSG antibodies, produces a colored reaction product. The amount of color developed in a given time period is proportional to the amount of LSG protein present in the sample. Quantitative results typically are obtained by reference to a standard curve.


A competition assay can also be employed wherein antibodies specific to LSG are attached to a solid support and labeled LSG and a sample derived from the host are passed over the solid support. The amount of label detected which is attached to the solid support can be correlated to a quantity of LSG in the sample.


Using all or a portion of a nucleic acid sequence of LSG of the present invention as a hybridization probe, nucleic acid methods can also be used to detect LSG mRNA as a marker for lung cancer. Polymerase chain reaction (PCR) and other nucleic acid methods, such as ligase chain reaction (LCR) and nucleic acid sequence based amplification (NASBA), can be used to detect malignant cells for diagnosis and monitoring of various malignancies. For example, reverse-transcriptase PCR (RT-PCR) is a powerful technique which can be used to detect the presence of a specific mRNA population in a complex mixture of thousands of other mRNA species. In RT-PCR, an mRNA species is first reverse transcribed to complementary DNA (cDNA) with use of the enzyme reverse transcriptase; the cDNA is then amplified as in a standard PCR reaction. RT-PCR can thus reveal by amplification the presence of a single species of mRNA. Accordingly, if the mRNA is highly specific for the cell that produces it, RT-PCR can be used to identify the presence of a specific type of cell.


Hybridization to clones or oligonucleotides arrayed on a solid support (i.e. gridding) can be used to both detect the expression of and quantitate the level of expression of that gene. In this approach, a cDNA encoding the LSG gene is fixed to a substrate. The substrate may be of any suitable type including but not limited to glass, nitrocellulose, nylon or plastic. At least a portion of the DNA encoding the LSG gene is attached to the substrate and then incubated with the analyte, which may be RNA or a complementary DNA (cDNA) copy of the RNA, isolated from the tissue of interest. Hybridization between the substrate bound DNA and the analyte can be detected and quantitated by several means including but not limited to radioactive labeling or fluorescence labeling of the analyte or a secondary molecule designed to detect the hybrid. Quantitation of the level of gene expression can be done by comparison of the intensity of the signal from the analyte compared with that determined from known standards. The standards can be obtained by in vitro transcription of the target gene, quantitating the yield, and then using that material to generate a standard curve.


Of the proteomic approaches, 2D electrophoresis is a technique well known to those in the art. Isolation of individual proteins from a sample such as serum is accomplished using sequential separation of proteins by different characteristics usually on polyacrylamide gels. First, proteins are separated by size using an electric current. The current acts uniformly on all proteins, so smaller proteins move farther on the gel than larger proteins. The second dimension applies a current perpendicular to the first and separates proteins not on the basis of size but on the specific electric charge carried by each protein. Since no two proteins with different sequences are identical on the basis of both size and charge, the result of a 2D separation is a square gel in which each protein occupies a unique spot. Analysis of the spots with chemical or antibody probes, or subsequent protein microsequencing can reveal the relative abundance of a given protein and the identity of the proteins in the sample.


The above tests can be carried out on samples derived from a variety of cells, bodily fluids and/or tissue extracts such as homogenates or solubilized tissue obtained from a patient. Tissue extracts are obtained routinely from tissue biopsy and autopsy material. Bodily fluids useful in the present invention include blood, urine, saliva or any other bodily secretion or derivative thereof. By blood it is meant to include whole blood, plasma, serum or any derivative of blood.


In Vivo Targeting of LSG/Lung Cancer Therapy


Identification of this LSG is also useful in the rational design of new therapeutics for imaging and treating cancers, and in particular lung cancer. For example, in one embodiment, antibodies which specifically bind to LSG can be raised and used in vivo in patients suspected of suffering from lung cancer. Antibodies which specifically bind LSG can be injected into a patient suspected of having lung cancer for diagnostic and/or therapeutic purposes. Thus, another aspect of the present invention provides for a method for preventing the onset and treatment of lung cancer in a human patient in need of such treatment by administering to the patient an effective amount of antibody. By “effective amount” it is meant the amount or concentration of antibody needed to bind to the target antigens expressed on the tumor to cause tumor shrinkage for surgical removal, or disappearance of the tumor. The binding of the antibody to the overexpriessed LSG is believed to cause the death of the cancer cell expressing such LSG. The preparation and use of antibodies for in vivo diagnosis and treatment is well known in the art. For example, antibody-chelators labeled with Indium-111 have been described for use in the radioimmunoscintographic imaging of carcinoembryonic antigen expressing tumors (Sumerdon et al. Nucl. Med. Biol. 1990 17:247-254). In particular, these antibody-chelators have been used in detecting tumors in patients suspected of having recurrent colorectal cancer (Griffin et al. J. Clin. Onc. 1991 9:631-640). Antibodies with paramagnetic ions as labels for use in magnetic resonance imaging have also been described (Lauffer, R. B. Magnetic Resonance in Medicine 1991 22:339-342). Antibodies directed against LSG can be used in a similar manner. Labeled antibodies which specifically bind LSG can be injected into patients suspected of having lung cancer for the purpose of diagnosing or staging of the disease status of the patient. The label used will be selected in accordance with the imaging modality to be used. For example, radioactive labels such as Indium-111, Technetium-99m or Iodine-131 can be used for planar scans or single photon emission computed tomography (SPECT). Positron emitting labels such as Fluorine-19 can be used in positron emission tomography. Paramagnetic ions such as Gadlinium (III) or Manganese (II) can be used in magnetic resonance imaging (MRI). Presence of the label, as compared to imaging of normal tissue, permits determination of the spread of the cancer. The amount of label within an organ or tissue also allows determination of the presence or absence of cancer in that organ or tissue.


Antibodies which can be used in in vivo methods include polyclonal, monoclonal and omniclonal antibodies and antibodies prepared via molecular biology techniques. Antibody fragments and aptamers and single-stranded oligonucleotides such as those derived from an in vitro evolution protocol referred to as SELEX and well known to those skilled in the art can also be used.


Screening Assays


The present invention also provides methods for identifying modulators which bind to LSG protein or have a modulatory effect on the expression or activity of LSG protein. Modulators which decrease the expression or activity of LSG protein are believed to be useful in treating lung cancer. Such screening assays are known to those of skill in the art and include, without limitation, cell-based assays and cell free assays.


Small molecules predicted via computer imaging to specifically bind to regions of LSG can also be designed, synthesized and tested for use in the imaging and treatment of lung cancer. Further, libraries of molecules can be screened for potential anticancer agents by assessing the ability of the molecule to bind to the LSGs identified herein. Molecules identified in the library as being capable of binding to LSG are key candidates for further evaluation for use in the treatment of lung cancer. In a preferred embodiment, these molecules will downregulate expression and/or activity of LSG in cells.


Adoptive Immunotherapy and Vaccines


Adoptive immunotherapy of cancer refers to a therapeutic approach in which immune cells with an antitumor reactivity are administered to a tumor-bearing host, with the aim that the cells mediate either directly or indirectly, the regression of an established tumor. Transfusion of lymphocytes, particularly T lymphocytes, falls into this category and investigators at the National Cancer Institute (NCI) have used autologous reinfusion of peripheral blood lymphocytes or tumor-infiltrating lymphocytes (TIL), T cell cultures from biopsies of subcutaneous lymph nodules, to treat several human cancers (Rosenberg, S. A., U.S. Pat. No. 4,690,914, issued Sep. 1, 1987; Rosenberg, S. A., et al., 1988, N. England J. Med. 319:1676-1680).


The present invention relates to compositions and methods of adoptive immunotherapy for the prevention and/or treatment of primary and metastatic lung cancer in humans using macrophages sensitized to the antigenic LSG molecules, with or without non-covalent complexes of heat shock protein (hsp). Antigenicity or immunogenicity of the LSG is readily confirmed by the ability of the LSG protein or a fragment thereof to raise antibodies or educate naive effector cells, which in turn lyse target cells expressing the antigen (or epitope).


Cancer cells are, by definition, abnormal and contain proteins which should be recognized by the immune system as foreign since they are not present in normal tissues. However, the immune system often seems to ignore this abnormality and fails to attack tumors. The foreign LSG proteins that are produced by the cancer cells can be used to reveal their presence. The LSG is broken into short fragments, called tumor antigens, which are displayed on the surface of the cell. These tumor antigens are held or presented on the cell surface by molecules called MHC, of which there are two types: class I and II. Tumor antigens in association with MHC class I molecules are recognized by cytotoxic T cells while antigen-MHC class II complexes are recognized by a second subset of T cells called helper cells. These cells secrete cytokines which slow or stop tumor growth and help another type of white blood cell, B cells, to make antibodies against the tumor cells.


In adoptive immunotherapy, T cells or other antigen presenting cells (APCs) are stimulated outside the body (ex vivo), using the tumor specific LSG antigen. The stimulated cells are then reinfused into the patient where they attack the cancerous cells. Research has shown that using both cytotoxic and helper T cells is far more effective than using either subset alone. Additionally, the LSG antigen may be complexed with heat shock proteins to stimulate the APCs as described in U.S. Pat. No. 5,985,270.


The APCs can be selected from among those antigen presenting cells known in the art including, but not limited to, macrophages, dendritic cells, B lymphocytes, and a combination thereof, and are preferably macrophages. In a preferred use, wherein cells are autologous to the individual, autologous immune cells such as lymphocytes, macrophages or other APCs are used to circumvent the issue of whom to select as the donor of the immune cells for adoptive transfer. Another problem circumvented by use of autologous immune cells is graft versus host disease which can be fatal if unsuccessfully treated.


In adoptive immunotherapy with gene therapy, DNA of the LSG can be introduced into effector cells similarly as in conventional gene therapy. This can enhance the cytotoxicity of the effector cells to tumor cells as they have been manipulated to produce the antigenic protein resulting in improvement of the adoptive immunotherapy.


LSG antigens of this invention are also useful as components of lung cancer vaccines. The vaccine comprises an immunogenically stimulatory amount of a LSG antigen. Immunogenically stimulatory amount refers to that amount of antigen that is able to invoke the desired immune response in the recipient for the amelioration, or treatment of lung cancer. Effective amounts may be determined empirically by standard procedures well known to those skilled in the art.


The LSG antigen may be provided in any one of a number of vaccine formulations which are designed to induce the desired type of immune response, e.g., antibody and/or cell mediated. Such formulations are known in the art and include, but are not limited to, formulations such as those described in U.S. Pat. No. 5,585,103. Vaccine formulations of the present invention used to stimulate immune responses can also include pharmaceutically acceptable adjuvants.


Vectors, Host Cells, Expression


The present invention also relates to vectors which include polynucleotides of the present invention, host cells which are genetically engineered with vectors of the invention and the production of polypeptides of the invention by recombinant techniques.


Host cells can be genetically engineered to incorporate LSG polynucleotides and express LSG polypeptides of the present invention. For instance, LSG polynucleotides may be introduced into host cells using well known techniques of infection, transduction, transfection, transvection and transformation. The LSG polynucleotides may be introduced alone or with other polynucleotides. Such other polynucleotides may be introduced independently, co-introduced or introduced joined to the LSG polynucleotides of the invention.


For example, LSG polynucleotides of the invention may be transfected into host cells with another, separate, polynucleotide encoding a selectable marker, using standard techniques for co-transfection and selection in, for instance, mammalian cells. In this case, the polynucleotides generally will be stably incorporated into the host cell genome.


Alternatively, the LSG polynucleotide may be joined to a vector containing a selectable marker for propagation in a host. The vector construct may be introduced into host cells by the-aforementioned techniques. Generally, a plasmid vector is introduced as DNA in a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid. Electroporation also may be used to introduce LSG polynucleotides into a host. If the vector is a virus, it may be packaged in vitro or introduced into a packaging cell and the packaged virus may be transduced into cells. A wide variety of well known techniques conducted routinely by those of skill in the art are suitable for making LSG polynucleotides and for introducing LSG polynucleotides into cells in accordance with this aspect of the invention. Such techniques are reviewed at length in reference texts such as Sambrook et al., previously cited herein.


Vectors which may be used in the present invention include, for example, plasmid vectors, single- or double-stranded phage vectors, and single- or double-stranded RNA or DNA viral vectors. Such vectors may be introduced into cells as polynucleotides, preferably DNA, by well known techniques for introducing DNA and RNA into cells. The vectors, in the case of phage and viral vectors, also may be and preferably are introduced into cells as packaged or encapsidated virus by well known techniques for infection and transduction. Viral vectors may be replication competent or replication defective. In the latter case viral propagation generally will occur only in complementing host cells.


Preferred vectors for expression of polynucleotides and polypeptides of the present invention include, but are not limited to, vectors comprising cis-acting control regions effective for expression in a host operatively linked to the polynucleotide to be expressed. Appropriate trans-acting factors either are supplied by the host, supplied by a complementing vector or supplied by the vector itself upon introduction into the host.


In certain preferred embodiments in this regard, the vectors provide for specific expression. Such specific expression may be inducible expression or expression only in certain types of cells or both inducible and cell-specific. Particularly preferred among inducible vectors are vectors that can be induced to express by environmental factors that are easy to manipulate, such as temperature and nutrient additives. A variety of vectors suitable to this aspect of the invention, including constitutive and inducible expression vectors for use in prokaryotic and eukaryotic hosts, are well known and employed routinely by those of skill in the art.


The engineered host cells can be cultured in conventional nutrient media which may be modified as appropriate for, inter alia, activating promoters, selecting transforymants or amplifying genes. Culture conditions such as temperature, pH and the like, previously used with the host cell selected for expression, generally will be suitable for expression of LSG polypeptides of the present invention.


A great variety of expression vectors can be used to express LSG polypeptides of the invention. Such vectors include chromosomal, episomal and virus-derived vectors. Vectors may be derived from bacterial plasmids, from bacteriophage, from yeast episomes, from yeast chromosomal elements, from viruses such as baculoviruses, papova viruses, such as SV40, vaccinia viruses, adenoviruses, fowl pox viruses, pseudorabies viruses and retroviruses, and from combinations thereof such as those derived from plasmid and bacteriophage genetic elements, such as cosmids and phagemids. All may be used for expression in accordance with this aspect of the present invention. Generally, any vector suitable to maintain, propagate or express polynucleotides to express a polypeptide in a host may be used for expression in this regard.


The appropriate DNA sequence may be inserted into the vector by any of a variety of well-known and routine techniques. In general, a DNA sequence for expression is joined to an expression vector by cleaving the DNA sequence and the expression vector with one or more restriction endonucleases and then joining the restriction fragments together using T4 DNA ligase. Procedures for restriction and ligation that can be used to this end are well known and routine to those of skill. Suitable procedures in this regard, and for constructing expression vectors using alternative techniques, which also are well known and routine to those skill, are set forth in great detail in Sambrook et al. cited elsewhere herein.


The DNA sequence in the expression vector is operatively linked to appropriate expression control sequence(s), including, for instance, a promoter to direct mRNA transcription. Representative promoters include the phage lambda PL promoter, the E. coli lac, trp and tac promoters, the SV40 early and late promoters, and promoters of retroviral LTRs, to name just a few of the well-known promoters. It will be understood that numerous promoters not mentioned are also suitable for use in this aspect of the invention and are well known and readily may be employed by those of skill in the manner illustrated by the discussion and the examples herein.


In general, expression constructs will contain sites for transcription initiation and termination, and, in the transcribed region, a ribosome binding site for translation. The coding portion of the mature transcripts expressed by the constructs will include a translation initiating AUG at the beginning and a termination codon appropriately positioned at the end of the polypeptide to be translated.


In addition, the constructs may contain control regions that regulate as well as engender expression. Generally, in accordance with many commonly practiced procedures, such regions will operate by controlling transcription, such as repressor binding sites and enhancers, among others.


Vectors for propagation and expression generally will include selectable markers. Such markers also may be suitable for amplification or the vectors may contain additional markers for this purpose. In this regard, the expression vectors preferably contain one or more selectable marker genes to provide a phenotypic trait for selection of transformed host cells. Preferred markers include dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, and tetracycline or ampicillin resistance genes for culturing in E. coli and other bacteria.


The vector containing the appropriate DNA sequence as described elsewhere herein, as well as an appropriate promoter, and other appropriate control sequences, may be introduced into an appropriate host using a variety of well known-techniques suitable to expression therein of a desired polypeptide. Representative examples of appropriate hosts include bacterial cells, such as E. coli, Streptomyces and Salmonella typhimurium cells; fungal cells, such as yeast cells; insect cells such as Drosophila S2 and Spodoptera Sf9 cells; animal cells such as CHO, COS and Bowes melanoma cells; and plant cells. Hosts for a great variety of expression constructs are well known, and those of skill will be enabled by the present disclosure readily to select a host for expressing a LSG polypeptide in accordance with this aspect of the present invention.


More particularly, the present invention also includes recombinant constructs, such as expression constructs, comprising one or more of the sequences described above. The constructs comprise a vector, such as a plasmid or viral vector, into which such LSG sequence of the invention has been inserted. The sequence may be inserted in a forward or reverse orientation. In certain preferred embodiments in this regard, the construct further comprises regulatory sequences, including, for example, a promoter, operably linked to the sequence. Large numbers of suitable vectors and promoters are known to those of skill in the art, and there are many commercially available vectors suitable for use in the present invention.


The following vectors, which are commercially available, are provided by way of example. Among vectors preferred for use in bacteria are pQE70, pQE60 and pQE-9, available from Qiagen; pBS vectors, Phagescript vectors, Bluescript vectors, pNH8A, pNH16a, pNH18A, pNH46A, available from Stratagene; and ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 available from Pharmacia. Among preferred eukaryotic vectors are PWLNEO, pSV2CAT, pOG44, pXT1 and pSG available from Stratagene; and pSVK3, pBPV, PMSG and pSVL available from Pharmacia. These vectors are listed solely by way of illustration of the many commercially available and well known vectors that are available to those of skill in the art for use in accordance with this aspect of the present invention. It will be appreciated by those of skill in the art upon reading this disclosure that any other plasmid or vector suitable for introduction, maintenance, propagation and/or expression of a LSG polynucleotide or polypeptide of the invention in a host may be used in this aspect of the invention.


Promoter regions can be selected from any desired gene using vectors that contain a reporter transcription unit lacking a promoter region, such as a chloramphenicol acetyl transferase (“cat”) transcription unit, downstream of a restriction site or sites for introducing a candidate promoter fragment; i.e., a fragment that may contain a promoter. As is well known, introduction into the vector of a promoter-containing fragment at the restriction site upstream of the cat gene engenders production of CAT activity detectable by standard CAT assays. Vectors suitable to this end are well known and readily available. Two such vectors are pKK232-8 and pCM7. Thus, promoters for expression of LSG polynucleotides of the present invention include, not only well known and readily available promoters, but also promoters that readily may be obtained by the foregoing technique, using a reporter gene.


Among known bacterial promoters suitable for expression of polynucleotides and polypeptides in accordance with the present invention are the E. coli laci and lacZ promoters, the T3 and T7 promoters, the gpt promoter, the lambda PR, PL promoters and the trp promoter.


Among known eukaryotic promoters suitable in this regard are the CMV immediate early promoter, the HSV thymidine kinase promoter, the early and late SV40 promoters, the promoters of retroviral LTRs, such as those of the Rous sarcoma virus (“RSV”), and metallothionein promoters, such as the mouse metallothionein-I promoter.


Selection of appropriate vectors and promoters for expression in a host cell is a well known procedure and the requisite techniques for expression vector construction, introduction of the vector into the host and expression in the host are routine skills in the art.


The present invention also relates to host cells containing the above-described constructs. The host cell can be a higher eukaryotic cell, such as a mammalian cell, or a lower eukaryotic cell, such as a yeast cell. Alternatively, the host cell can be a prokaryotic cell, such as a bacterial cell.


Introduction of the construct into the host cell can be effected by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection or other methods. Such methods are described in many standard laboratory manuals, such as Davis et al. BASIC METHODS IN MOLECULAR BIOLOGY, (1986).


Constructs in host cells can be used in a conventional manner to produce the gene product encoded by the recombinant sequence. Alternatively, LSG polypeptides of the invention can be synthetically produced by conventional peptide synthesizers.


Mature proteins can be expressed in mammalian cells, yeast, bacteria, or other cells under the control of appropriate promoters. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention. Appropriate cloning and expression vectors for use with prokaryotic and eukaryotic hosts are described by Sambrook et al. cited elsewhere herein.


Generally, recombinant expression vectors will include origins of replication, a promoter derived from a highly-expressed gene to direct transcription of a downstream structural sequence, and a selectable marker to permit isolation of vector containing cells after exposure to the vector. Among suitable promoters are those derived from the genes that encode glycolytic enzymes such as 3-phosphoglycerate kinase (“PGK”), a-factor, acid phosphatase, and heat shock proteins, among others. Selectable markers include the ampicillin resistance gene of E. coli and the trpl gene of S. cerevisiae.


Transcription of DNA encoding the LSG polypeptides of the present invention by higher eukaryotes may be increased by inserting an enhancer sequence into the vector. Enhancers are cis-acting elements of DNA, usually about from to 300 base pairs (bp) that act to increase transcriptional activity of a promoter in a given host cell-type. Examples of enhancers include the SV40 enhancer, which is located on the late side of the replication origin at bp 100 to 270, the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers.


A polynucleotide of the present invention, encoding a heterologous structural sequence of a LSG polypeptide of the present invention, generally will be inserted into the vector using standard techniques so that it is operably linked to the promoter for expression. The polynucleotide will be positioned so that the transcription start site is located appropriately 5′ to a ribosome binding site. The ribosome binding site will be 5′ to the AUG that initiates translation of the polypeptide to be expressed. Generally, there will be no other open reading frames that begin with an initiation codon, usually AUG, lying between the ribosome binding site and the initiating AUG. Also, generally, there will be a translation stop codon at the end of the polypeptide and there will be a polyadenylation signal and a transcription termination signal appropriately disposed at the 3′ end of the transcribed region.


Appropriate secretion signals may be incorporated into the expressed polypeptide for secretion of the translated protein into the lumen of the endoplasmic reticulum, into the periplasmic space or into the extracellular environment. The signals may be endogenous to the polypeptide or they may be heterologous signals.


The polypeptide may be expressed in a modified form, such as a fusion protein, and may include not only secretion signals but also additional heterologous functional regions. Thus, for instance, a region of additional amino acids,, particularly charged amino acids, may be added to the N-terminus of the polypeptide to improve stability and persistence in the host cell during purification or during subsequent handling and storage. A region also may be added to the polypeptide to facilitate purification. Such regions may be removed prior to final preparation of the polypeptide. The addition of peptide moieties to polypeptides to engender secretion or excretion, to improve stability and to facilitate purification, among others, are familiar and routine techniques in the art.


Suitable prokaryotic hosts for propagation, maintenance or expression of LSG polynucleotides and polypeptides in accordance with the invention include Escherichia coli, Bacillus subtilis and Salmonella typhimurium. Various species of Pseudomonas, Streptomyces, and Staphylococcus are suitable hosts in this regard. Many other hosts also known to those of skill may also be employed in this regard.


As a representative, but non-limiting example, useful expression vectors for bacterial use can comprise a selectable marker and bacterial origin of replication derived from commercially available plasmids comprising genetic elements of the well known cloning vector pBR322. Such commercial vectors include, for example, pKK223-3 (Pharmacia Fine Chemicals, Uppsala, Sweden) and GEM1 (Promega Biotec, Madison, Wis., USA). These pBR322 “backbone” sections are combined with an appropriate promoter and the structural sequence to be expressed. Following transformation of a suitable host strain and growth of the host strain to an appropriate cell density, where the selected promoter is inducible it is induced by appropriate means (e.g., temperature shift or exposure to chemical inducer) and cells are cultured for an additional period. Cells typically then are harvested by centrifugation, disrupted by physical or chemical means, and the resulting crude extract retained for further purification. Microbial cells employed in expression of proteins can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents, such methods are well know to those skilled in the art.


Various mammalian cell culture systems can be employed for expression, as well. An exemplary mammalian expression systems is the COS-7 line of monkey kidney fibroblasts described in Gluzman et al., Cell 23: 175 (1981). Other mammalian cell lines capable of expressing a compatible vector include for example, the C127, 3T3, CHO, HeLa, human kidney 293 and BHK cell lines. Mammalian expression vectors comprise an origin of replication, a suitable promoter and enhancer, and any ribosome binding sites, polyadenylation sites, splice donor and acceptor sites, transcriptional termination sequences, and 5′ flanking non-transcribed sequences that are necessary for expression. In certain preferred embodiments in this regard DNA sequences derived from the SV40 splice sites, and the SV40 polyadenylation sites are used for required non-transcribed genetic elements of these types.


LSG polypeptides can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Most preferably, high performance liquid chromatography (“HPLC”) is employed for purification. Well known techniques for refolding proteins may be employed to regenerate active conformation when the polypeptide is denatured during isolation and or purification.


LSG polypeptides of the present invention include naturally purified products, products of chemical synthetic procedures, and products produced by recombinant techniques from a prokaryotic or eukaryotic host, including, for example, bacterial, yeast, higher plant, insect and mammalian cells. Depending upon the host employed in a recombinant production procedure, the LSG polypeptides of the present invention may be glycosylated or may be non-glycosylated. In addition, LSG polypeptides of the invention may also include an initial modified methionine residue, in some cases as a result of host-mediated processes.


LSG polynucleotides and polypeptides may be used in accordance with the present invention for a variety of applications, particularly those that make use of the chemical and biological properties of the LSGs. Additional applications relate to diagnosis and to treatment of disorders of cells, tissues and organisms. These aspects of the invention are illustrated further by the following discussion.


Polynucleotide Assays


As discussed in some detail supra, this invention is also related to the use of LSG polynucleotides to detect complementary polynucleotides such as, for example, as a diagnostic reagent. Detection of a mutated form of LSG associated with a dysfunction will provide a diagnostic tool that can add to or define a diagnosis of a disease or susceptibility to a disease which results from under-expression, over-expression or altered expression of a LSG, such as, for example, a susceptibility to inherited lung cancer.


Individuals carrying mutations in a human LSG gene may be detected at the DNA level by a variety of techniques. Nucleic acids for diagnosis may be obtained from a patient's cells, such as from blood, urine, saliva, tissue biopsy and autopsy material. The genomic DNA may be used directly for detection or may be amplified enzymatically using PCR prior to analysis(Saiki et al., Nature, 324: 163-166 (1986)). RNA or cDNA may also be used in a similar manner. As an example, PCR primers complementary to a LSG polynucleotide of SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74 can be used to identify and analyze LSG expression and mutations. For example, deletions and insertions can be detected by a change in size of the amplified product in comparison to the normal genotype. Point mutations can be identified by hybridizing amplified DNA to radiolabeled LSG RNA or alternatively, radiolabeled LSG antisense DNA sequences. Perfectly matched sequences can be distinguished from mismatched duplexes by RNase A digestion or by differences in melting temperatures.


Sequence differences between a reference gene and genes having mutations also may be revealed by direct DNA sequencing. In addition, cloned DNA segments may be employed as probes to detect specific DNA segments. The sensitivity of such methods can be greatly enhanced by appropriate use of PCR or another amplification method. For example, a sequencing primer is used with double-stranded PCR product or a single-stranded template molecule generated by a modified PCR. The sequence determination is performed by conventional procedures with radiolabeled nucleotide or by automatic sequencing procedures with fluorescent-tags.


Genetic testing based on DNA sequence differences may be achieved by detection of alterations in electrophoretic mobility of DNA fragments in gels, with or without denaturing agents. Small sequence deletions and insertions can be visualized by high resolution gel electrophoresis. DNA fragments of different sequences may be distinguished on denaturing formamide gradient gels in which the mobilities of different DNA fragments are retarded in the gel at different positions according to their specific melting or partial melting temperatures. (see, e.g., Myers et al., Science, 230: 1242 (1985)).


Sequence changes at specific locations also may be revealed by nuclease protection assays, such as RNase and S1 protection or the chemical cleavage method (e.g., Cotton et al., Proc. Natl. Acad. Sci., USA, 85: 4397-4401 (1985)).


Thus, the detection of a specific DNA sequence may be achieved by methods such as hybridization, RNase protection, chemical cleavage, direct DNA sequencing or the use of restriction enzymes, (e.g., restriction fragment length polymorphisms (“RFLP”) and Southern blotting of genomic DNA. In addition to more conventional gel-electrophoresis and DNA sequencing, mutations also can be detected by in situ analysis.


Chromosome Assays


The LSG sequences of the present invention are also valuable for chromosome identification. There is a need for identifying particular sites on the chromosome and few chromosome marking reagents based on actual sequence data (repeat polymorphisms) are presently available for marking chromosomal location. Each LSG sequence of the present invention is specifically targeted to and can hybridize with a particular location on an individual human chromosome. Thus, the LSGs can be used in the mapping of DNAs to chromosomes, an important first step in correlating sequences with genes associated with disease.


In certain preferred embodiments in this regard, the cDNA herein disclosed is used to clone genomic DNA of a LSG of the present invention. This can be accomplished using a variety of well known techniques and libraries, which generally are available commercially. The genomic DNA is used for in situ chromosome mapping using well known techniques for this purpose.


In some cases, sequences can be mapped to chromosomes by preparing PCR primers (preferably 15-25 bp) from the cDNA. Computer analysis of the 3′ untranslated region of the gene is used to rapidly select primers that do not span more than one exon in the genomic DNA, thus complicating the amplification process. These primers are then used for PCR screening of somatic cell hybrids containing individual human chromosomes. Only those hybrids containing the human gene corresponding to the primer will yield an amplified fragment.


PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular DNA to a particular chromosome. Using the present invention with the same oligonucleotide primers, sublocalization can be achieved with panels of fragments from specific chromosomes or pools of large genomic clones in an analogous manner. Other mapping strategies that can similarly be used to map to its chromosome include in situ hybridization, prescreening with labeled flow-sorted chromosomes and preselection by hybridization to construct chromosome specific-cDNA libraries.


Fluorescence in situ hybridization (“FISH”) of a cDNA clone to a metaphase chromosomal spread can be used to provide a precise chromosomal location in one step. This technique can be used with cDNA as short as 50 or 60 bp. This technique is described by Verma et al. (HUMAN CHROMOSOMES: A MANUAL OF BASIC TECHNIQUES, Pergamon Press, New York (1988)).


Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be correlated with genetic map data. Such data are found, for example, in V. McKusick, MENDELIAN INHERITANCE IN MAN, available on line through Johns Hopkins University, Welch Medical Library. The relationship between genes and diseases that have been mapped to the same chromosomal region are then identified through linkage analysis (coinheritance of physically adjacent genes).


Next, it is necessary to determine the differences in the cDNA or genomic sequence between affected and unaffected individuals. If a mutation is observed in some or all of the affected individuals but not in any normal individuals, then the mutation is likely to be the causative agent of the disease.


With current resolution of physical mapping and genetic mapping techniques, a cDNA precisely localized to a chromosomal region associated with the disease could be one of between 50 and 500 potential causative genes. (This assumes 1 megabase mapping resolution and one gene per 20 kb).


Polypeptide Assays


As described in some detail supra, the present invention also relates to diagnostic assays such as quantitative and diagnostic assays for detecting levels of LSG polypeptide in cells and tissues, and biological fluids such as blood and urine, including determination of normal and abnormal levels. Thus, for instance, a diagnostic assay in accordance with the present invention for detecting over-expression or under-expression of a LSG polypeptide compared to normal control tissue samples may be used to detect the presence of neoplasia. Assay techniques that can be used to determine levels of a protein, such as a LSG polypeptide of the present invention, in a sample derived from a host are well-known to those of skill in the art. Such assay methods include radioimmunoassays, competitive-binding assays, Western Blot analysis and ELISA assays. Among these ELISAs frequently are preferred.


For example, antibody-sandwich ELISAs are used to detect polypeptides in a sample, preferably a biological sample. Wells of a microtiter plate are coated with specific antibodies, at a final concentration of 0.2 to 10 μg/ml. The antibodies are either monoclonal or polyclonal and are produced by methods as described herein. The wells are blocked so that non-specific binding of the polypeptide to the well is reduced. The coated wells are then incubated for >2 hours at room temperature with a sample containing the LSG polypeptide. Preferably, serial dilutions of the sample should be used to validate results. The plates are then washed three times with deionized or distilled water to remove unbounded polypeptide. Next, 50 μl of specific antibody-alkaline phosphatase conjugate, at a concentration of 25-400 ng, is added and incubated for 2 hours at room temperature. The plates are again washed three times with deionized or distilled water to remove unbounded conjugate. 4-methylumbelliferyl phosphate (MUP) or p-nitrophenyl phosphate (NPP) substrate solution (75 μl) is then added to each well and the plate is incubated 1 hour at room temperature. The reaction is measured by a microtiter plate reader. A standard curve is prepared using serial dilutions of a control sample, and polypeptide concentration is plotted on the X-axis (log scale) while fluorescence or absorbance is plotted on the Y-axis (linear scale). The concentration of the LSG polypeptide in the sample is interpolated using the standard curve.


Antibodies


As discussed in some detail supra, LSG polypeptides, their fragments or other derivatives, or analogs thereof, or cells expressing them can be used as an immunogen to produce antibodies thereto. These antibodies can be polyclonal or monoclonal antibodies. The present invention also includes chimeric, single chain, and humanized antibodies, as well as Fab fragments, or the product of an Fab expression library. Various procedures known in the art may be used for the production of such antibodies and fragments.


A variety of methods for antibody production are set forth in Current Protocols, Chapter 2.


For example, cells expressing a LSG polypeptide of the present invention can be administered to an animal to induce the production of sera containing polyclonal antibodies. In a preferred method, a preparation of the secreted protein is prepared and purified to render it substantially free of natural contaminants. This preparation is then introduced into an animal in order to produce polyclonal antisera of greater specific activity. The antibody obtained will bind with the LSG polypeptide itself. In this manner, even a sequence encoding only a fragment of the LSG polypeptide can be used to generate antibodies binding the whole native polypeptide. Such antibodies can then be used to isolate the LSG polypeptide from tissue expressing that LSG polypeptide.


Alternatively, monoclonal antibodies can be prepared. Examples of techniques for production of monoclonal antibodies include, but are not limited to, the hybridoma technique (Kohler, G. and Milstein, C., Nature 256: 495-497 (1975), the trioma technique, the human B-cell hybridoma technique (Kozbor et al., Immunology Today 4: 72 (1983) and (Cole et al., pg. 77-96 in MONOCLONAL ANTIBODIES AND CANCER THERAPY, Alan R. Liss, Inc. (1985). The EBV-hybridoma technique is useful in production of human monoclonal antibodies.


Hybridoma technologies have also been described by Khler et al. (Eur. J. Immunol. 6: 511 (1976)) Khler et al. (Eur. J.Immunol. 6: 292 (1976)) and Hammerling et al. (in: Monoclonal Antibodies and T-Cell Hybridomas, Elsevier, N.Y., pp. 563-681 (1981)). In general, such procedures involve immunizing an animal (preferably a mouse) with LSG polypeptide or, more preferably, with a secreted LSG polypeptide-expressing cell. Such cells may be cultured in any suitable tissue culture medium; however, it is preferable to culture cells in Earle's modified Eagle's medium supplemented with 10% fetal bovine serum (inactivated at about 56° C.), and supplemented with about 10 g/l of nonessential amino acids, about 1,000 U/ml of penicillin, and about 100 μg/ml of streptomycin. The splenocytes of such mice are extracted and fused with a suitable myeloma cell line. Any suitable myeloma cell line may be employed in accordance with the present invention; however, it is preferable to employ the parent myeloma cell line (SP20), available from the ATCC. After fusion, the resulting hybridoma cells are selectively maintained in HAT medium, and then cloned by limiting dilution as described by Wands et al. (Gastroenterology 80: 225-232 (1981).). The hybridoma cells obtained through such a selection are then assayed to identify clones which secrete antibodies capable of binding the polypeptide.


Alternatively, additional antibodies capable of binding to the polypeptide can be produced in a two-step procedure using anti-idiotypic antibodies. Such a method makes use of the fact that antibodies are themselves antigens, and therefore, it is possible to obtain an antibody which binds to a second antibody. In accordance with this method, protein specific antibodies are used to immunize an animal, preferably a mouse. The splenocytes of such an animal are then used to produce hybridoma cells, and the hybridoma cells are screened to identify clones which produce an antibody whose ability to bind to the protein-specific antibody can be blocked by the polypeptide. Such antibodies comprise anti-idiotypic antibodies to the protein specific antibody and can be used to immunize an animal to induce formation of further protein-specific antibodies.


Techniques described for the production of single chain antibodies (U.S. Pat. No. 4,946,778) can also be adapted to produce single chain antibodies to immunogenic polypeptide products of this invention. Also, transgenic mice, as well as other nonhuman transgenic animals, may be used to express humanized antibodies to immunogenic polypeptide products of this invention.


It will be appreciated that Fab, F(ab′)2 and other fragments of the antibodies of the present invention may also be used according to the methods disclosed herein. Such fragments are typically produced by proteolytic cleavage, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab′)2 fragments). Alternatively, secreted protein-binding fragments can be produced through the application of recombinant DNA technology or through synthetic chemistry.


For in vivo use of antibodies in humans, it may be preferable to use “humanized” chimeric monoclonal antibodies. Such antibodies can be produced using genetic constructs derived from hybridoma cells producing the monoclonal antibodies described above. Methods for producing chimeric antibodies are known in the art (See, for review, Morrison, Science 229: 1202 (1985); Oi et al., BioTechniques 4: 214 (1986); Cabilly et al., U.S. Pat. No. 4,816,567; Taniguchi et al., EP 171496; Morrison et al., EP 173494; Neuberger et al., WO 8601533; Robinson et al., WO 8702671; Boulianne et al., Nature 312: 643 (1984); Neuberger et al., Nature 314: 268 (1985).)


The above-described antibodies may be employed to isolate or to identify clones expressing LSG polypeptides or purify LSG polypeptides of the present invention by attachment of the antibody to a solid support for isolation and/or purification by affinity chromatography. As discussed in more detail supra, antibodies specific against a LSG may also be used to image tumors, particularly cancer of the lung, in patients suffering from cancer. Such antibodies may also be used therapeutically to target tumors expressing a LSG.


Preferred exemplary antigenic epitopes of LSGs of the present invention which have been identified are depicted below. The antigenicity index (AI avg) used is Jameson-Wolf. In some embodiment, it may be preferred to raise antibodies against these regions of the LSGs.

Antigenicity Index(Jameson-Wolf)positionsAI avglengthDEX73_2.aa(SEQ ID NO: 75)16-501.0635DEX73_3.aa(SEQ ID NO: 76)52-661.0515DEX73_5.aa(SEQ ID NO: 77)1419-14331.16151387-14141.0828808-8251.0018DEX73_8.aa(SEQ ID NO: 78)208-2231.0616123-1351.0513689-7171.032963-901.0228653-6831.0131366-3771.0012DEX73_12.aa(SEQ ID NO: 80)56-681.0513DEX73_13.aa(SEQ ID NO: 81)207-2171.281172-851.1714405-4691.1565151-1711.0221DEX73_18.aa(SEQ ID NO: 84)40-511.2712


LSG Binding Molecules and Assays


This invention also provides a method for identification of molecules, such as receptor molecules, that bind LSGs. Genes encoding proteins that bind LSGs, such as receptor proteins, can be identified by numerous methods known to those of skill in the art. Examples include, but are not limited to, ligand panning and FACS sorting. Such methods are described in many laboratory manuals such as, for instance, Coligan et al., Current Protocols in Immunology 1(2): Chapter 5 (1991).


Expression cloning may also be employed for this purpose. To this end, polyadenylated RNA is prepared from a cell responsive to a LSG of the present invention. A cDNA library is created from this RNA and the library is divided into pools. The pools are then transfected individually into cells that are not responsive to a LSG of the present invention. The transfected cells then are exposed to labeled LSG. LSG polypeptides can be labeled by a variety of well-known techniques including, but not limited to, standard methods of radio-iodination or inclusion of a recognition site for a site-specific protein kinase. Following exposure, the cells are fixed and binding of labeled LSG is determined. These procedures conveniently are carried out on glass slides. Pools containing labeled LSG are identified as containing cDNA that produced LSG-binding cells. Sub-pools are then prepared from these positives, transfected into host cells and screened as described above. Using an iterative sub-pooling and re-screening process, one or more single clones that encode the putative binding molecule, such as a receptor molecule, can be isolated.


Alternatively a labeled ligand can be photoaffinity linked to a cell extract, such as a membrane or a membrane extract, prepared from cells that express a molecule that it binds, such as a receptor molecule. Cross-linked material is resolved by polyacrylamide gel electrophoresis (“PAGE”) and exposed to X-ray film. The labeled complex containing the ligand-receptor can be excised, resolved into peptide fragments, and subjected to protein microsequencing. The amino acid sequence obtained from microsequencing can be used to design unique or degenerate oligonucleotide probes to screen cDNA libraries to identify genes encoding the putative receptor molecule.


Polypeptides of the invention also can be used to assess LSG binding capacity of LSG binding molecules, such as receptor molecules, in cells or in cell-free preparations.


Agonists and Antagonists—Assays and Molecules


The invention also provides a method of screening compounds to identify those which enhance or block the action of a LSG on cells. By “compound”, as used herein, it is meant to be inclusive of small organic molecules, peptides, polypeptides and antibodies as well as any other candidate molecules which have the potential to enhance or agonize or block or antagonize the action of LSG on cells. As used herein, an agonist is a compound which increases the natural biological functions of a LSG or which functions in a manner similar to a LSG, while an antagonist, as used herein, is a compound which decreases or eliminates such functions. Various known methods for screening for agonists and/or antagonists can be adapted for use in identifying LSG agonist or antagonists.


For example, a cellular compartment, such as a membrane or a preparation thereof, such as a membrane-preparation, may be prepared from a cell that expresses a molecule that binds a LSG, such as a molecule of a signaling or regulatory pathway modulated by LSG. The preparation is incubated with labeled LSG in the absence or the presence of a compound which may be a LSG agonist or antagonist. The ability of the compound to bind the binding molecule is reflected in decreased binding of the labeled ligand. Compounds which bind gratuitously, i.e., without inducing the effects of a LSG upon binding to the LSG binding molecule are most likely to be good antagonists. Compounds that bind well and elicit effects that are the same as or closely related to LSG are agonists. LSG-like effects of potential agonists and antagonists may by measured, for instance, by determining activity of a second messenger system following interaction of the candidate molecule with a cell or appropriate cell preparation, and comparing the effect with that of LSG or molecules that elicit the same effects as LSG. Second messenger systems that may be useful in this regard include, but are not limited to, AMP guanylate cyclase, ion channel or phosphoinositide hydrolysis second messenger systems.


Another example of an assay for LSG antagonists is a competitive assay that combines LSG and a potential antagonist with membrane-bound LSG receptor molecules or recombinant LSG receptor molecules under appropriate conditions for a competitive inhibition assay. LSG can be labeled, such as by radioactivity, such that the number of LSG molecules bound to a receptor molecule can be determined accurately to assess the effectiveness of the potential antagonist.


Potential antagonists include small organic molecules, peptides, polypeptides and antibodies that bind to a LSG polypeptide of the invention and thereby inhibit or extinguish its activity. Potential antagonists also may be small organic molecules, a peptide, a polypeptide such as a closely related protein or antibody that binds the same sites on a binding molecule, such as a receptor molecule, without inducing LSG-induced activities, thereby preventing the action of LSG by excluding LSG from binding.


Potential antagonists include small molecules which bind to and occupy the binding site of the LSG polypeptide thereby preventing binding to cellular binding molecules, such as receptor molecules, such that normal biological activity is prevented. Examples of small molecules include but are not limited to small organic molecules, peptides or peptide-like molecules.


Other potential antagonists include antisense molecules. Antisense technology can be used to control gene expression through antisense DNA or RNA or through triple-helix formation. Antisense techniques are discussed, for example, in Okano, J. Neurochem. 56: 560 (1991); OLIGODEOXYNUCLEOTIDES AS ANTISENSE INHIBITORS OF GENE EXPRESSION, CRC Press, Boca Raton, Fla. (1988). Triple helix formation is discussed in, for instance Lee et al., Nucleic Acids Research 6: 3073 (1979); Cooney et al., Science 241: 456 (1988); and Dervan et al., Science 251: 1360 (1991). The methods are based on binding of a polynucleotide to a complementary DNA or RNA. For example, the 5′ coding portion of a polynucleotide that encodes a mature LSG polypeptide of the present invention may be used to design an antisense RNA oligonucleotide of from about 10 to 40 base pairs in length. A DNA oligonucleotide is designed to be complementary to a region of the gene involved in transcription thereby preventing transcription and the production of a LSG polypeptide. The antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into a LSG polypeptide. The oligonucleotides described above can also be delivered to cells such that the antisense RNA or DNA may be expressed in vivo to inhibit production of a LSG.


Compositions


The present invention also relates to compositions comprising a LSG polynucleotide or a LSG polypeptide or an agonist or antagonist thereof.


For example, a LSG polynucleotide, polypeptide or an agonist or antagonist thereof of the present invention may be employed in combination with a non-sterile or sterile carrier or carriers for use with cells, tissues or organisms, such as a pharmaceutical carrier suitable for administration to a subject. Such compositions comprise, for instance, a media additive or a therapeutically effective amount of a polypeptide of the invention and a pharmaceutically acceptable carrier or excipient. Such carriers may include, but are not limited to, saline, buffered saline, dextrose, water, glycerol, ethanol and combinations thereof. The formulation should suit the mode of administration.


Compositions of the present invention will be formulated and dosed in a fashion consistent with good medical practice, taking into account the clinical condition of the individual patient (especially the side effects of treatment with the polypeptide or other compound alone), the site of delivery, the method of administration, the scheduling of administration, and other factors known to practitioners. The “effective amount” for purposes herein is thus determined by such considerations.


As a general proposition, the total pharmaceutically effective amount of secreted polypeptide administered parenterally per dose will be in the range of about 1, μg/kg/day to 10 mg/kg/day of patient body weight, although, as noted above, this will be subject to therapeutic discretion. More preferably, this dose is at least 0.01 mg/kg/day, and most preferably for humans between about 0.01 and 1 mg/kg/day for the hormone. If given continuously, the polypeptide or other compound is typically administered at a dose rate of about 1 μg/kg/hour to about 50 mg/kg/hour, either by 1-4 injections per day or by continuous subcutaneous infusion, for example, using a mini-pump. An intravenous bag solution may also be employed. The length of treatment needed to observe changes and the interval following treatment for responses to occur appears to vary depending on the desired effect.


Pharmaceutical compositions containing the secreted protein of the invention are administered orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, gels, drops or transdermal patch), bucally, or as an oral or nasal spray. “Pharmaceutically acceptable carrier” refers to a non-toxic solid, semisolid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type. The term “parenteral” as used herein refers to modes of administration which include intravenous, intramuscular, intraperitoneal, intrasternal, subcutaneous and intraarticular injection and infusion.


The polypeptide or other compound is also suitably administered by sustained-release systems. Suitable examples of sustained-release compositions include semipermeable polymer matrices in the form of shaped articles, e. g., films, or microcapsules. Sustained-release matrices include polylactides (U.S. Pat. No. 3,773,919 and EP 58481), copolymers of L-glutamic acid and gamma-ethyl-L-glutamate (Sidman, U. et al., Biopolymers 22: 547-556 (1983)), poly (2-hydroxyethyl methacrylate) (R. Langer et al., J. Biomed. Mater. Res. 15: 167-277 (1981), and R. Langer, Chem. Tech. 12: 98-105 (1982)), ethylene vinyl acetate (R. Langer et al.) and poly-D-(−)-3-hydroxybutyric acid (EP 133,988). Sustained-release compositions also include liposomally entrapped polypeptides. Liposomes containing the polypeptide or other compound are prepared by well known methods (Epstein et al., Proc. Natl. Acad. Sci. USA 82: 3688-3692 (1985); Hwang et al., Proc. Natl. Acad. Sci. USA 77: 4030-4034 (1980); EP 52322; EP 36676; EP 88046; EP 143949; EP 142641; Japanese Pat. Appl. 83-118008; U.S. Pat. Nos. 4,485,045 and 4,544,545; and EP 102324). Ordinarily, the liposomes are of the small (about 200-800 Angstroms) unilamellar type in which the lipid content is greater than about 30 mol. percent cholesterol, the selected proportion being adjusted for the optimal therapy.


For parenteral administration, in one embodiment, the polypeptide or other compound is formulated generally by mixing it at the desired degree of purity, in a unit dosage injectable form (solution, suspension, or emulsion), with a pharmaceutically acceptable carrier, i.e., one that is non-toxic to recipients at the dosages and concentrations employed and is compatible with other ingredients of the formulation.


For example, the formulation preferably does not include oxidizing agents and other compounds that are known to be deleterious to the polypeptide or other compound.


Generally, the formulations are prepared by contacting the polypeptide or other compound uniformly and intimately with liquid carriers or finely divided solid carriers or both. Then, if necessary, the product is shaped into the desired formulation. Preferably the carrier is a parenteral carrier, more preferably a solution that is isotonic with the blood of the recipient. Examples of such carrier vehicles include water, saline, Ringer's solution, and dextrose solution. Non-aqueous vehicles such as fixed oils and ethyl oleate are also useful herein, as well as liposomes.


The carrier suitably contains minor amounts of additives such as substances that enhance isotonicity and chemical stability. Such materials are non-toxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, succinate, acetic acid, and other organic acids or their salts; antioxidants such as ascorbic acid; low molecular weight (less than about ten residues) polypeptides, e. g., polyarginine or tripeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids, such as glycine, glutamic acid, aspartic acid, or arginine; monosaccharides, disaccharides, and other carbohydrates including cellulose or its derivatives, glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; counterions such as sodium; and/or nonionic surfactants such as polysorbates, poloxamers, or PEG.


The polypeptide or other compound is typically formulated in such vehicles at a concentration of about 0.1 mg/ml to 100 mg/ml, preferably 1-10 mg/ml, at a pH of about 3 to 8. It will be understood that the use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of polypeptide salts or salts of the other compounds.


Any polypeptide to be used for therapeutic administration should be sterile. Sterility is readily accomplished by filtration through sterile filtration membranes (e. g., 0.2 micron membranes). Therapeutic polypeptide compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.


Polypeptides ordinarily will be stored in unit or multi-dose containers, for example, sealed ampules or vials, as an aqueous solution or as a lyophilized formulation for reconstitution. As an example of a lyophilized formulation, 10-ml vials are filled with 5 ml of sterile-filtered 1% (w/v) aqueous polypeptide solution, and the resulting mixture is lyophilized. The infusion solution is prepared by reconstituting the lyophilized polypeptide using bacteriostatic Water-for-Injection.


Kits


The invention further relates to pharmaceutical packs and kits comprising one or more containers filled with one or more of the ingredients of the aforementioned compositions of the invention. Associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, reflecting approval by the agency of the manufacture, use or sale of the product for human administration.


Administration


LSG polypeptides or polynucleotides or-other compounds, preferably agonists or antagonists thereof of the present invention may be employed alone or in conjunction with other compounds, such as therapeutic compounds.


The pharmaceutical compositions may be administered in any effective, convenient manner including, for instance, administration by topical, oral, anal, vaginal, intravenous, intraperitoneal, intramuscular, subcutaneous, intranasal or intradermal routes among others.


The pharmaceutical compositions generally are administered in an amount effective for treatment or prophylaxis of a specific indication or indications. In general, the compositions are administered in an amount of at least about 10 μg/kg body weight. However, it will be appreciated that optimum dosage will be determined by standard methods for each treatment modality and indication, taking into account the indication, its severity, route of administration, complicating conditions and the like.


It will be appreciated that conditions caused by a decrease in the standard or normal expression level of a LSG polypeptide in an individual can be treated by administering the LSG polypeptide of the present invention, preferably in the secreted form, or an agonist thereof. Thus, the invention also provides a method of treatment of an individual in need of an increased level of a LSG polypeptide comprising administering to such an individual a pharmaceutical composition comprising an amount of the LSG polypeptide or an agonist thereof to increase the activity level of the LSG polypeptide in such an individual. For example, a patient with decreased levels of a LSG polypeptide may receive a daily dose 0.1-100 μg/kg of a LSG polypeptide or agonist thereof for six consecutive days. Preferably, if a LSG polypeptide is administered it is in the secreted form.


Compositions of the present invention can also be administered to treating increased levels of a LSG polypeptide. For example, antisense technology can be used to inhibit production of a LSG polypeptide of the present invention. This technology is one example of a method of decreasing levels of a polypeptide, preferably a secreted form, due to a variety of etiologies, such as cancer. A patient diagnosed with abnormally increased levels of a polypeptide can be administered intravenously antisense polynucleotides at 0.5, 1.0, 1.5, 2.0 and 3.0 mg/kg day for 21 days. This treatment is preferably repeated after a 7-day rest period if the treatment was well tolerated. Compositions comprising an antagonist of a LSG polypeptide can also be administered to decrease levels of LSG in a patient.


Gene Therapy


The LSG polynucleotides, polypeptides, agonists and antagonists that are polypeptides may be employed in accordance with the present invention by expression of such polypeptides in vivo, in treatment modalities often referred to as “gene therapy.”


Thus, for example, cells from a patient may be engineered with a polynucleotide, such as a DNA or RNA, encoding a polypeptide ex vivo, and the engineered cells then can be provided to a patient to be treated with the polypeptide. For example, cells may be engineered ex vivo by the use of a retroviral plasmid vector containing RNA encoding a polypeptide of the present invention. Such methods are well-known in the art and their use in the present invention will be apparent from the teachings herein.


Similarly, cells may be engineered in vivo for expression of a polypeptide in vivo by procedures known in the art. For example, a polynucleotide of the invention may be engineered for expression in a replication defective retroviral vector, as discussed supra. The retroviral expression construct then may be isolated and introduced into a packaging cell transduced with a retroviral plasmid vector containing RNA encoding a polypeptide of the present invention such that the packaging cell now produces infectious viral particles containing the gene of interest. These producer cells may be administered to a patient for engineering cells in vivo and expression of the polypeptide in vivo. These and other methods for administering a polypeptide of the present invention would be apparent to those skilled in the art upon reading the instant application.


Retroviruses from which the retroviral plasmid vectors herein above mentioned may be derived include, but are not limited to, Moloney Murine Leukemia Virus, spleen necrosis virus, retroviruses such as Rous Sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, gibbon ape leukemia virus, human immunodeficiency virus, adenovirus, Myeloproliferative Sarcoma Virus, and mammary tumor virus. In one embodiment, the retroviral plasmid vector is derived from Moloney Murine Leukemia Virus.


Such vectors will include one or more promoters for expressing the polypeptide. The selection of a suitable promoter will be apparent to those skilled in the art from the teachings contained herein. However, examples of suitable promoters which may be employed include, but are not limited to, the retroviral LTR, the SV40 promoter, the human cytomegalovirus (CMV) promoter described in Miller et al., Biotechniques 7: 980-990 (1989), and eukaryotic cellular promoters such as the histone, RNA polymerase III, and beta-actin promoters. Other viral promoters which may be employed include, but are not limited to, adenovirus promoters, thymidine kinase (TK) promoters, and B19 parvovirus promoters. Additional promoters which may be used include respiratory syncytial virus (RSV) promoter, inducible promoters such as the MMT promoter, the metallothionein promoter, heat shock promoters, the albumin promoter, the ApoAI promoter, human globin promoters, viral thymidine kinase promoters such as the Herpes Simplex thymidine kinase promoter, retroviral LTRs, the beta-actin promoter, and human growth hormone promoters. The promoter also may be the native promoter which controls the gene encoding the polypeptide.


The nucleic acid sequence encoding the polypeptide of the present invention will be placed under the control of a suitable promoter.


In one embodiment, the retroviral plasmid vector is employed to transduce packaging cell lines to form producer cell lines. Examples of packaging cells which may be transfected include, but are not limited to, the PE501, PA317, Y-2, Y-AM, PA12, T19-14X, VT-19-17-H2, YCRE, YCRIP, GP+E-86, GP+envAm12, and DAN cell lines as described in Miller, A., Human Gene Therapy 1: 5-14 (1990). The vector may be transduced into the packaging cells through any means known in the art. Such means include, but are not limited to, electroporation, the use of liposomes, and CaPO4 precipitation. Alternatively, the retroviral plasmid vector may be encapsulated into a liposome, or coupled to a lipid, and then administered to a host. The producer cell line will generate infectious retroviral vector particles which are inclusive of the nucleic acid sequence(s) encoding the polypeptides. Such retroviral vector particles then may be employed to transduce eukaryotic cells, either in vitro or in vivo. The transduced eukaryotic cells will express the nucleic acid sequence(s) encoding the polypeptide. Eukaryotic cells which may be transduced include, but are not limited to, embryonic stem cells, embryonic carcinoma cells, as well as hematopoietic stem cells, hepatocytes, fibroblasts, myoblasts, keratinocytes, endothelial cells, and bronchial epithelial cells.


An exemplary method of gene therapy involves transplantation of fibroblasts which are capable of expressing a LSG polypeptide or an agonist or antagonist thereof onto a patient. Generally fibroblasts are obtained from a subject by skin biopsy. The resulting tissue is placed in tissue-culture medium and separated into small pieces. Small chunks of the tissue are placed on a wet surface of a tissue culture flask, approximately ten pieces are placed in each flask. The flask is turned upside down, closed tight and left at room temperature over night. After 24 hours at room temperature, the flask is inverted and the chunks of tissue remain fixed to the bottom of the flask and fresh media (e. g., Ham's F12 media, with 10% FBS, penicillin and streptomycin) is added. The flasks are then incubated at 37° C. for approximately one week. At this time, fresh media is added and subsequently changed every several days. After an additional two weeks in culture, a monolayer of fibroblasts emerge. The monolayer is trypsinized and scaled into larger flasks. pMV-7 (Kirschmeier, P. T. et al., DNA, 7: 219-25 (1988)), flanked by the long terminal repeats of the Moloney murine sarcoma virus, is digested with EcoRI and HindIII and subsequently treated with calf intestinal phosphatase. The linear vector is fractionated on agarose gel and purified, using glass beads. The cDNA encoding a LSG polypeptide of the present invention or an agonist or antagonist thereof can be amplified using PCR primers which correspond to their 5′ and 3′ end sequences respectively. Preferably, the 5′ primer contains an EcoRI site and the 3′ primer includes a HindIII site. Equal quantities of the Moloney murine sarcoma virus linear backbone and the amplified EcoRI and HindIII fragment are added together in the presence of T4 DNA ligase. The resulting mixture is maintained under conditions appropriate for ligation of the two fragments. The ligation mixture is then used to transform bacteria HB 101, which are then plated onto agar containing kanamycin for the purpose of confirming that the vector has the gene of interest properly inserted. Amphotropic pA317 or GP+am12 packaging cells are grown in tissue culture-to confluent density in Dulbeccols Modified Eagles Medium (DMEM) with 10% calf serum (CS), penicillin and streptomycin. The MSV vector containing the gene is then added to the media and the packaging cells transduced with the vector. The packaging cells now produce infectious viral particles containing the gene (the packaging cells are now referred to as producer cells). Fresh media is added to the transduced producer cells, and subsequently, the media is harvested from a 10 cm plate of confluent producer cells. The spent media, containing the infectious viral particles, is filtered through a millipore filter to remove detached producer cells and this media is then used to infect fibroblast cells. Media is removed from a sub-confluent plate of fibroblasts and quickly replaced with the media from the producer cells. This media is removed and replaced with fresh media. If the titer of virus is high, then virtually all fibroblasts will be infected and no selection is required. If the titer is very low, then it is necessary to use a retroviral vector that has a selectable marker, such as neo or his. Once the fibroblasts have been efficiently infected, the fibroblasts are analyzed to determine whether protein is produced. The engineered fibroblasts are then transplanted onto the host, either alone or after having been grown to confluence on cytodex 3 microcarrier beads.


Alternatively, in vivo gene therapy methods can be used to treat LSG related disorders, diseases and conditions. Gene therapy methods relate to the introduction of naked nucleic acid (DNA, RNA, and antisense DNA or RNA) sequences into an animal to increase or decrease the expression of the polypeptide.


For example, a LSG polynucleotide of the present invention or a nucleic acid sequence encoding an agonist or antagonist thereto may be operatively linked to a promoter or any other genetic elements necessary for the expression of the polypeptide by the target tissue. Such gene therapy and delivery techniques and methods are known in the art, see, for example, WO 90/11092, WO 98/11779; U.S. Pat. Nos. 5,693,622, 5,705,151, and 5,580,859; Tabata H. et al. (1997) Cardiovasc. Res. 35 (3): 470-479, Chao J et al. (1997) Pharmacol. Res. 35 (6): 517-522, Wolff J. A. (1997) Neuromuscul. Disord. 7 (5): 314-318, Schwartz B. et al. (1996) Gene Ther. 3 (5): 405-411, Tsurumi Y. et al. (1996) Circulation 94 (12): 3281-3290 (incorporated herein by reference). The polynucleotide constructs may be delivered by any method that delivers injectable materials to the cells of an animal, such as, injection into the interstitial space of tissues (heart, muscle, skin, lung, liver, intestine and the like). The polynucleotide constructs can be delivered in a pharmaceutically acceptable liquid or aqueous carrier.


The term “naked” polynucleotide, DNA or RNA, refers to sequences that are free from any delivery vehicle that acts to assist, promote, or facilitate entry into the cell, including viral sequences, viral particles, liposome formulations, lipofectin or precipitating agents and the like. However, polynucleotides may also be delivered in liposome formulations (such as those taught in Felgner P. L. et al. (1995) Ann. NY Acad. Sci. 772: 126-139 and Abdallah B. et al. (1995) Biol. Cell 85 (1): 1-7) which can be prepared by methods well known to those skilled in the art.


The polynucleotide vector constructs used in the gene therapy method are preferably constructs that will not integrate into the host genome nor will they contain sequences that allow for replication. Any strong promoter known to those skilled in the art can be used for driving the expression of DNA. Unlike other gene therapies techniques, one major advantage of introducing naked nucleic acid sequences into target cells is the transitory nature of the polynucleotide synthesis in the cells. Studies have shown that non-replicating DNA sequences can be introduced into cells to provide production of the desired polypeptide for periods of up to six months.


The polynucleotide construct can be delivered to the interstitial space of tissues within the an animal, including of muscle, skin, brain, lung, liver, spleen, bone marrow, thymus, heart, lymph, blood, bone, cartilage, pancreas, kidney, gall bladder, stomach, intestine, testis, ovary, uterus, rectum, nervous system, eye, gland, and connective tissue. Interstitial space of the tissues comprises the intercellular fluid, mucopolysaccharide matrix among the reticular fibers of organ tissues, elastic fibers in the walls of vessels or chambers, collagen fibers of fibrous tissues, or that same matrix within connective tissue ensheathing muscle cells or in the lacunae of bone. It is similarly the space occupied by the plasma of the circulation and the lymph fluid of the lymphatic channels. Delivery to the interstitial space of muscle tissue is preferred. The polynucleotide construct may be conveniently delivered by injection into the tissues comprising these cells. They are preferably delivered to and expressed in persistent, non-dividing cells which are differentiated, although delivery and expression may be achieved in non-differentiated or less completely differentiated cells, such as, for example, stem cells of blood or skin fibroblasts. In vivo muscle cells are particularly competent in their ability to take up and express polynucleotides.


For the naked polynucleotide injection, an effective dosage amount of DNA or RNA will be in the range of from about 0.05 μg/kg body weight to about 50 mg/kg body weight. Preferably the dosage will be from about 0.005 mg/kg to about 20 mg/kg and more preferably from about 0.05 mg/kg to about 5 mg/kg. Of course, as the artisan of ordinary skill will appreciate, this dosage will vary according to the tissue site of injection. The appropriate and effective dosage of nucleic acid sequence can readily be determined by those of ordinary skill in the art and may depend on the condition being treated and the route of administration. The preferred route of administration is by the parenteral route of injection into the interstitial space of tissues. However, other parenteral routes may also be used, such as, inhalation of an aerosol formulation particularly for delivery to lungs or bronchial tissues, throat or mucous membranes of the nose. In addition, naked polynucleotide constructs can be delivered to arteries during angioplasty by the catheter used in the procedure.


The dose response effects of injected polynucleotide in muscle in vivo is determined as follows. Suitable template DNA for production of mRNA coding for polypeptide of the present invention is prepared in accordance with a standard recombinant DNA methodology. The template DNA, which may be either circular or linear, is either used as naked DNA or complexed with liposomes. The quadriceps muscles of mice are then injected with various amounts of the template DNA.


Five to six week old female and male Balb/C mice are anesthetized by intraperitoneal injection with 0.3 ml of 2.5% Avertin. A 1.5 cm incision is made on the anterior thigh, and the quadriceps muscle is directly visualized. The template DNA is injected in 0.1 ml of carrier in a 1 cc syringe through a 27 gauge needle over one minute, approximately 0.5 cm from the distal insertion site of the muscle into the knee and about 0.2 cm deep. A suture is placed over the injection site for future localization, and the skin is closed with stainless steel clips.


After an appropriate incubation time (e.g., 7 days) muscle extracts are prepared by excising the entire quadriceps. Every fifth 15 μm cross-section of the individual quadriceps muscles is histochemically stained for protein expression. A time course for protein expression may be done in a similar fashion except that quadriceps from different mice are harvested at different times. Persistence of DNA in muscle following injection may be determined by Southern blot analysis after preparing total cellular DNA and HIRT supernatants from injected and control mice.


The results of the above experimentation in mice can be use to extrapolate proper dosages and other treatment parameters in humans and other animals using naked DNA.


Nonhuman Transgenic Animals


The LSG polypeptides of the invention can also be expressed in nonhuman transgenic animals. Nonhuman animals of any species, including, but not limited to, mice, rats, rabbits, hamsters, guinea pigs, pigs, micro-pigs, goats, sheep, cows and non-human primates, e. g., baboons, monkeys, and chimpanzees, may be used to generate transgenic animals. Any technique known in the art may be used to introduce the transgene (I. e., polynucleotides of the invention) into animals to produce the founder lines of transgenic animals. Such techniques include, but are not limited to, pronuclear microinjection (Paterson et al., Appl. Microbiol. Biotechnol. 40: 691-698 (1994); Carver et al., Biotechnology (NY) 11: 1263-1270 (1993); Wright et al., Biotechnology (NY) 9: 830-834 (1991); and Hoppe et al., U.S. Pat. No. 4,873,191); retrovirus mediated gene transfer into germ lines (Van der Putten et al., Proc. Natl. Acad. Sci., USA 82: 6148-6152 (1985)), blastocysts or embryos; gene targeting in embryonic stem cells (Thompson et al., Cell 56: 313-321 (1989)); electroporation of cells or embryos (Lo, 1983, Mol. Cell. Biol. 3: 1803-1814 (1983)); introduction of the polynucleotides of the invention using a gene gun (see, e.g., Ulmer et al., Science 259: 1745 (1993); introducing nucleic acid constructs into embryonic pluripotent stem cells and transferring the stem cells back into the blastocyst; and sperm mediated gene transfer (Lavitrano et al., Cell 57: 717-723 (1989)). For a review of such techniques, see Gordon,“Transgenic Animals,” Intl. Rev. Cytol. 115: 171-229 (1989), which is incorporated by reference herein in its entirety.


Any technique known in the art may be used to produce transgenic clones containing polynucleotides of the invention, for example, nuclear transfer into enucleated oocytes of nuclei from cultured embryonic, fetal, or adult cells induced to quiescence (Campell et al., Nature 380: 64-66 (1996); Wilmut et al., Nature 385: 810813 (1997)).


The present invention provides for transgenic animals that carry the transgene in all their cells, as well as animals which carry the transgene in some, but not all their cells, i.e., mosaic or chimeric animals. The transgene may be integrated as a single transgene or as multiple copies such as in concatamers, e. g., head-to-head tandems or head-to-tail tandems. The transgene may also be selectively introduced into and activated in a particular cell type by following, for example, the teaching of Lasko et al. (Lasko et al., Proc. Natl. Acad. Sci. USA 89: 6232-6236 (1992)). The regulatory sequences required for such a cell-type specific activation will depend upon the particular cell type of interest, and will be apparent to those of skill in the art. When it is desired that the polynucleotide transgene be integrated into the chromosomal site of the endogenous gene, gene targeting is preferred. Briefly, when such a technique is to be utilized, vectors containing some nucleotide sequences homologous to the endogenous gene are designed for the purpose of integrating, via homologous recombination with chromosomal sequences, into and disrupting the function of the nucleotide sequence of the endogenous gene. The transgene may also be selectively introduced into a particular cell type, thus inactivating the endogenous gene in only that cell type, by following, for example, the teaching of Gu et al. (Science 265: 103-106 (1994)). The regulatory sequences required for such a cell-type specific inactivation will depend upon the particular cell type of interest, and will be apparent to those of skill in the art.


Once transgenic animals have been generated, the expression of the recombinant gene may be assayed utilizing standard techniques. Initial screening may be accomplished by Southern blot analysis or PCR techniques to analyze animal tissues to verify that integration of the transgene has taken place. The level of mRNA expression of the transgene in the tissues of the transgenic animals may also be assessed using techniques which include, but are not limited to, Northern blot analysis of tissue samples obtained from the animal, in situ hybridization analysis, and reverse transcriptase-PCR (rt-PCR). Samples of transgenic gene-expressing tissue may also be evaluated immunocytochemically or immunohistochemically using antibodies specific for the transgene product.


Once the founder animals are produced, they may be bred, inbred, outbred, or crossbred to produce colonies of the particular animal. Examples of such breeding strategies include, but are not limited to: outbreeding of founder animals with more than one integration site in order to establish separate lines; inbreeding of separate lines in order to produce compound transgenics that express the transgene at higher levels because of the effects of additive expression of each transgene; crossing of heterozygous transgenic animals to produce animals homozygous for a given integration site in order to both augment expression and eliminate the need for screening of animals by DNA analysis; crossing of separate homozygous lines to produce compound heterozygous or homozygous lines; and breeding to place the transgene on a distinct background that is appropriate for an experimental model of interest.


Transgenic animals of the invention have uses which include, but are not limited to, animal model systems useful in elaborating the biological function of LSG polypeptides of the present invention, studying conditions and/or disorders associated with aberrant expression of LSGS, and in screening for compounds effective in ameliorating such LSG associated conditions and/or disorders.


Knock-Out Animals


Endogenous gene expression can also be reduced by inactivating or “knocking out” the gene and/or its promoter using targeted homologous recombination (e. g., see Smithies et al., Nature 317: 230-234 (1985); Thomas & Capecchi, Cell 51: 503512 (1987); Thompson et al., Cell 5:



313-321 (1989); each of which is incorporated by reference herein in its entirety). For example, a mutant, non-functional LSG polynucleotide of the invention (or a completely unrelated DNA sequence) flanked by DNA homologous to the endogenous LSG polynucleotide sequence (either the coding regions or regulatory regions of the gene) can be used, with or without a selectable marker and/or a negative selectable marker, to transfect cells that express polypeptides of the invention in vivo. In another embodiment, techniques known in the art are used to generate knockouts in cells that contain, but do not express the gene of interest. Insertion of the DNA construct, via targeted homologous recombination, results in inactivation of the targeted gene. Such approaches are particularly suited in research and agricultural fields where modifications to embryonic stem cells can be used to generate animal offspring with an inactive targeted gene (e. g., see Thomas & Capecchi 1987 and Thompson 1989, supra). This approach can also be routinely adapted for use in humans provided the recombinant DNA constructs are directly administered or targeted to the required site in vivo using appropriate viral vectors that will be apparent to those of skill in the art.


In further embodiments of the invention, cells that are genetically engineered to express the LSG polypeptides of the invention, or alternatively, that are genetically engineered not to express the LSG polypeptides of the invention (e. g., knockouts) are administered to a patient in vivo. Such cells may be obtained from the patient or a MHC compatible donor and can include, but are not limited to, fibroblasts, bone marrow cells, blood cells (e. g., lymphocytes), adipocytes, muscle cells, and endothelial cells. The cells are genetically engineered in vitro using recombinant DNA techniques to introduce the coding sequence of polypeptides of the invention into the cells, or alternatively, to disrupt the coding sequence and/or endogenous regulatory sequence associated with the polypeptides of the invention, e. g., by transduction (using viral vectors, and preferably vectors that integrate the transgene into the cell genome) or transfection procedures, including, but not limited to, the use of plasmids, cosmids, YACs, naked DNA, electroporation, liposomes, etc.


The coding sequence of the LSG polypeptides of the invention can be placed under the control of a strong constitutive or inducible promoter or promoter/enhancer to achieve expression, and preferably secretion, of the LSG polypeptides of the invention. The engineered cells which express and preferably secrete the LSG polypeptides of the invention can be introduced into the patient systemically, e.g., in the circulation, or intraperitoneally.


Alternatively, the cells can be incorporated into a matrix and implanted in the body, e.g., genetically engineered fibroblasts can be implanted as part of a skin graft or genetically engineered endothelial cells can be implanted as part of a lymphatic or vascular graft (see, for example, U.S. Pat. No. 5,399,349 and U.S. Pat. No. 5,460,959 each of which is incorporated by reference herein in its entirety).


When the cells to be administered are non-autologous or non-MHC compatible cells, they can be administered using well known techniques which prevent the development of a host immune response against the introduced cells. For example, the cells may be introduced in an encapsulated form which, while allowing for an exchange of components with the immediate extracellular environment, does not allow the introduced cells to be recognized by the host immune system.


Transgenic and “knock-out” animals of the invention have uses which include, but are not limited to, animal model systems useful in elaborating the biological function of LSG polypeptides of the present invention, studying conditions and/or disorders associated with aberrant LSG expression, and in screening for compounds effective in ameliorating such LSG associated conditions and/or disorders.


The following nonlimiting example is provided to further illustrate the present invention.


EXAMPLE

The following Example is carried out using standard techniques, which are well known and routine to those of skill in the art, except where otherwise described in detail. Routine molecular biology techniques of the following example can be carried out as described in standard laboratory manuals, such as Sambrook et al., MOLECULAR CLONING: A LABORATORY MANUAL, 2nd Ed.; Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989).


Introduction and Background for Microarray Analysis


cDNA microarrays are prepared by high-speed robotic printing of thousands of distinct cDNAs in an ordered array on glass microscope slides. They are used to measure the relative abundance of specific sequences in two complex samples (Schena et al, 1995; Shalon et al, 1996).


In the microarray procedure, mRNA is isolated from tissues of interest, either from a tumor or control (normal or normal adjacent tissue). mRNA (200-600 ng) from cancer tissue or control is reverse transcribed to incorporate the fluorescent nucleotides Cy5 (red) or Cy3 (green), respectively. The two populations of fluorescently labeled cDNA are mixed together and hybridized simultaneously to a microarray bearing approximately 10,000 cDNA elements in a 2 cm×2 cm area on a glass slide (Microarrays hybridization service: Incyte Genomics, Fremont, Calif., USA). After hybridization, the slides are scanned with a scanning laser confocal microscope.


The scanned image is used to generate the intensity and local background measurements for each spot on the array (GEMtools software, Incyte Genomics). For each spot, representing one EST, the ratio of the normalized Cy5/Cy3 intensities generates a quantitation of the gene's expression in one tissue relative to the control, in this case, the expression in cancer tissue versus either normal or normal adjacent tissue. For example, a gene that shows a Cancer-Cy5 intensity of 3000 and a Normal-Cy3 intensity of 1000 is expressed 3-fold more in cancer tissue. Advanced analysis software is used to sort and decipher patterns of gene expression from the data (Cluster and Treeview programs, Stanford University; Eisen et al, 1998; Alizadeh et al, 2000). However, the reproducibility study from Incyte shows that the level of detectable differential expression is calculated to be approximately plus or minus 1.74. Consequently, any elements with observed ratios greater than or equal to 1.8 between cancer and normal are deemed differentially expressed.


References:




  • 1. Schena, M., D. Shalon, R. W. Davis, and P. O. Brown. 1995. Quantitative monitoring of gene expression patterns with a complementary cDNA microarray. Science 270: 467-470.

  • 2. Shalon, D., S. J. Smith, and P. O. Brown. 1996. A DNA Microarray System for Analyzing Complex DNA samples Using Two-color Fluorescent Probe Hybridization. Genome Research 6: 639-645.

  • 3. Eisen, M. B., P. T. Spellman, P. O. Brown, and D. Botstein. 1998. “Cluster analysis and display of genome-wide expression patterns”. PNAS 95: 14863-14868.

  • 4. Alizadeh, A. A., et al, 2000. “Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling.” Nature, 403: 503-511.

  • 5. GEM Microarray Reproducibility Study. Technical specifications from Incyte Genomics.


    Lung DiaDexus Microarray Candidates



Following is a list of “diaDexus microarray candidates” sequences for lung cancer, also referred to herein as lung specific genes or LSGs:

ddx QPCRSEQ IDGene IDddx lung codelung code1/19979057Lng128Lng1282/20347842Lng129Lng129 3983439Lng112Lng112 4236582Lng114Lng114 5210995Lng118Lng118 6208994Lng121Lng121 71066498Lng124Lng124 8287016Lng126Lng126 910717SQLng001Lng1361024945SQLng006Lng1431152017SQLng007Lng14412460254SQLng110Lng13813/74179090SQLng012Lng137146348SQLng004Lng1421594694SQLng005Lng14016145812SQLng008Lng1511710713SQLng002Lng1501820152SQLng003Lng141


Example 1

Sequence 1


Lng128


Gene ID 979057

TABLE 1The absolute numbers are relative levels of expression ofLng128 in 24 normal different tissues. All the values arecompared to normal trachea (calibrator). These RNA samplesare commercially pools, originated by pooling samples ofa particular tissue from different individuals.TissueNORMALAdrenal Gland0.03Bladder0.00Brain6.68Cervix0.00Colon0.00Endometrium0.12Esophagus0.00Heart0.01Kidney0.02Liver0.03Lung35.63Mammary Gland0.02Muscle0.00Ovary1.11Pancreas17.94Prostate0.42Rectum0.16Small Intestine0.00Spleen1.27Stomach0.00Testis2.17Thymus0.13Trachea1.00Uterus0.09
0 = negative


The relative levels of expression in Table 1 show that Lng128 mRNA expression is much higher in lung (35.63) compared with most other normal tissues analyzed.


The absolute numbers in Table 1 were obtained analyzing pools of samples of a particular tissue from different individuals. They can not be compared to the absolute numbers originated from RNA obtained from tissue samples of a single individual in Table 2.

TABLE 2The absolute numbers are relative levels of expression of Lng128 in 69 pairsof matching samples and 1 ovary normal and one ovary cancer sample. All thevalues are compared to normal trachea (calibrator). A matching pair is formedby mRNA from the cancer sample for a particular tissue and mRNA from the normaladjacent sample for that same tissue from the same individual.MATCHINGNORMALSample IDCancer TypeTissueCANCERADJACENTLng 60LAdenocarcinomaLung 1196.0417.03Lng 143LAdenocarcinomaLung 221.930.88Lng 60XLAdenocarcinomaLung 317.2142.37Lng AC82AdenocarcinomaLung 443.264.56Lng AC88AdenocarcinomaLung 5364.56101.48Lng AC66AdenocarcinomaLung 617.9413.27Lng AC69AdenocarcinomaLung 7582.0539.12Lng AC11AdenocarcinomaLung 824.42113.38Lng AC32AdenocarcinomaLung 9648.0727.19Lng AC39AdenocarcinomaLung 10249.001.71Lng AC94AdenocarcinomaLung 1142.81112.99Lng AC90AdenocarcinomaLung 12196.720.48Lng 47XQAdenocarcinomaLung 1388.950.54Lng 223LAdenocarcinomaLung 145.800.00Lng 528LAdenocarcinomaLung 1545.25177.91Lng BR26Bronchio-alveolarLung 162.8028.54carcinomaLng BA641BronchogenicLung 171746.2036.13carcinomaLng 315LSquamous cellLung 181.67736.73carcinomaLng SQ45Squamous cellLung 19828.8762.68carcinomaLng SQ14Squamous cellLung 200.0715.56carcinomaLng SQ9XSquamous cellLung 2173.264.32carcinomaLng SQ56Squamous cellLung 2233.24141.53carcinomaLng SQ80Squamous cellLung 23101.1344.79carcinomaLng SQ32Squamous cellLung 24119.439.82carcinomaLng SQ16Squamous cellLung 2564.0010.85carcinomaLng SQ79Squamous cellLung 2652.16142.52carcinomaLng 90XSquamous cellLung 2738.726.23carcinomaLng BR94Squamous cellLung 2827.190.00carcinomaLng C20XSquamous cellLung 290.001.59carcinomaLng SQ44Squamous cellLung 3013.880.04carcinomaLng SQ43Squamous cellLung 3124.001.39carcinomaLng 77LLarge cellLung 320.1513.93carcinomaLng LC71Large cellLung 3361.61190.68carcinomaLng LC109Large cellLung 3425.19513.78carcinomaLng LC80Large cellLung 35537.4547.01carcinomaLng 75XCMetastatic fromLung 3644.7939.95bone cancerLng MT71Metastatic fromLung 3711.3526.45renal cell cancerLng MT67Metastatic fromLung 383.287.97melanomaBld 46XKBladder 10.000.00Bld TR14Bladder 20.460.00Cvx KS52Cervix 10.290.00Cvx KS83Cervix 20.000.00Cln AS45Colon 10.000.00Cln RC01Colon 20.000.10End 8911Endometrium 10.080.68End 28XAEndometrium 212.730.57kid 107XDKidney 10.020.02Kid 109XDKidney 20.050.25Liv 94XALiver 10.000.00Liv 174LLiver 20.000.00Mam 162XMammary 10.000.02Mam 497MMammary 20.000.00Ovr A082Ovary 10.031.57Ovr 18GAOvary 25.78Ovr 180BOvary 30.03Pan 71XPancreas 10.030.02Pan 92XPancreas 20.650.00Pro 109XBProstate 10.010.03Pro 125XBProstate 20.020.02Skn 248SSkin 10.110.00Skn 816SSkin 21.010.00SmInt 21XASmall Intestine 10.010.00SmInt H89Small Intestine 20.672.76Sto 758SStomach 10.000.00Sto 531SStomach 20.080.00Tst 647TTestis 14.380.96Tst 39XTestis 28.691.19Thr 143NThyroid 10.150.00Thr 270TThyroid 20.000.00Utr 135XOUterus 10.190.27Utr 141XOUterus 20.060.00
0 = Negative


In the analysis of matching samples, higher expression of lng128 is detected in lung samples showing a high degree of tissue specificity for lung tissue. These results confirm the tissue specificity results obtained with normal pooled samples (Table 1).


Furthermore, we compared the level of mRNA expression in cancer samples and the isogenic normal adjacent tissue from the same individual. This comparison provides an indication of specificity for the cancer stage (e.g. higher levels of mRNA expression in the cancer sample compared to the normal adjacent). Table 2 shows overexpression of Lng128 in 38 lung cancer tissues compared with their respective normal adjacent (lung samples #1, 2, 4, 5, 7, 9, 10, 12, 13, 14, 17, 19, 21, 23, 24, 25, 27, 28, 30, 31, 35 and 35,). There is overexpression in the cancer tissue for 55% of the lung matching samples tested (21 out of total of 38 lung matching samples).


Altogether, the high level of tissue specificity, plus the mRNA differential expression in the lung matching samples tested are believed to make Lng128 a good marker for diagnosing, monitoring, staging, imaging and treating lung cancer.


Northern Analysis


Two transcripts ˜2.2 kb and ˜4.2 kb


Primers Used for QPCR Expression Analysis

Forward primerCTTGGTCTTCCTGCTCCTGAC(SEQ ID NO:21)Reverse primerAGGGCAGAGAGGAACAGCA(SEQ ID NO:22)ProbeCCAGCGAGGAGCAGCAGGGATG(SEQ ID NO:23)


Example 2

Sequence 2


Lng129


Gene ID 347842

TABLE 1The absolute numbers are relative levels of expression ofLng129 in 24 normal different tissues. All the values arecompared to normal spleen (calibrator). These RNA samplesare commercially available pools, originated by pooling samplesof a particular tissue from different individuals.TissueNORMALAdrenal Gland0.00Bladder0.00Brain0.00Cervix0.02Colon0.00Endometrium0.03Esophagus0.00Heart0.00Kidney0.00Liver0.01Lung0.12Mammary Gland0.00Muscle0.00Ovary0.04Pancreas0.00Prostate0.01Rectum0.00Small Intestine0.00Spleen1.00Stomach0.00Testis0.01Thymus0.03Trachea0.06Uterus0.06
0 = negative


The relative levels of expression in Table 1 show that Lng129 mRNA expression is high compared with most other normal tissues analyzed.


The absolute numbers in Table 1 were obtained analyzing pools of samples of a particular tissue from different individuals. They can not be compared to the absolute numbers originated from RNA obtained from tissue samples of a single individual in Table 2.

TABLE 2The absolute numbers are relative levels of expression of Lng129 in 67 pairsof matching samples and 1 ovary normal and one ovary cancer sample. All thevalues are compared to normal spleen (calibrator). A matching pair is formedby mRNA from the cancer sample for a particular tissue and mRNA from the normaladjacent sample for that same tissue from the same individual.CANCERSample IDCancer TypeTissueMATCHINGNORMALLng 60LAdenocarcinomaLung 10.710.69Lng 143LAdenocarcinomaLung 20.010.00Lng 60XLAdenocarcinomaLung 30.000.01Lng AC82AdenocarcinomaLung 40.310.00Lng AC88AdenocarcinomaLung 50.000.00Lng AC66AdenocarcinomaLung 60.800.07Lng AC69AdenocarcinomaLung 70.000.00Lng AC11AdenocarcinomaLung 80.480.06Lng AC32AdenocarcinomaLung 90.390.00Lng AC39AdenocarcinomaLung 100.530.01Lng AC94AdenocarcinomaLung 110.050.03Lng AC90AdenocarcinomaLung 120.040.00Lng 47XQAdenocarcinomaLung 130.120.00Lng 223LAdenocarcinomaLung 140.040.00Lng 528LAdenocarcinomaLung 150.000.00Lng BR26Bronchio-alveolarLung 160.240.94Lng BA641BronchogenicLung 170.400.10carcinomaLng 315LSquamous cellLung 180.030.12Lng SQ45Squamous cellLung 190.000.00Lng SQ14Squamous cellLung 200.020.11Lng SQ9XSquamous cellLung 210.000.00Lng SQ56Squamous cellLung 220.430.12Lng SQ80Squamous cellLung 230.000.00Lng SQ32Squamous cellLung 240.060.00Lng SQ16Squamous cellLung 250.010.00Lng SQ79Squamous cellLung 260.110.04Lng 90XSquamous cellLung 270.000.00Lng BR94Squamous cellLung 284.760.00Lng C20XSquamous cellLung 290.000.00Lng SQ44Squamous cellLung 300.040.00Lng SQ43Squamous cellLung 310.820.08Lng 77LLarge cellLung 320.000.00carcinomaLng LC71Large cellLung 330.050.30carcinomaLng LC109Large cellLung 341.480.90carcinomaLng LC80Large cellLung 351.090.00carcinomaLng 75XCMetastatic fromLung 360.000.00boneLng MT71Metastatic fromLung 370.180.04renalLng MT67Metastatic fromLung 380.550.04Bld 46XKBladder 10.020.00Bld TR14Bladder 20.460.39Cvx KS52Cervix 10.260.03Cvx KS83Cervix 20.000.00ClnAS45Colon 10.000.00ClnRC01Colon 20.010.02EndEndometrium0.000.00kid 107XDKidney 11.530.03Kid 109XDKidney 20.330.11Liv 175LLiver 10.270.03Liv174LLiver 20.010.01MamMammary 10.020.01Mam 497MMammary 20.000.00Ovr A082Ovary 10.010.00Ovr 18GAOvary 20.01Ovr 180BOvary 30.00Pan 77XPancreas 10.000.00PanPancreas 20.000.00ProProstate 10.010.02ProProstate 20.000.00Skn 248SSkin 10.130.02SmIntSmall0.020.01SmInt H89Small0.000.00StoStomach 10.150.01Sto 531SStomach 20.000.00Tst647TTestis 10.000.00Tst 39XTestis 20.300.02ThrThyroid 10.040.03Thr 270TThyroid 20.110.00Utr135XOUterus 10.200.00Utr 141XOUterus 20.000.01
0 = Negative


In the analysis of matching samples, higher expression of lng129 is detected in lung samples showing a high degree of tissue specificity for lung tissue. These results confirm the tissue specificity results obtained with normal pooled samples (Table 1).


Furthermore, we compared the level of mRNA expression in cancer samples and the isogenic normal adjacent tissue from the same individual. This comparison provides an indication of specificity for the cancer stage (e.g. higher levels of mRNA expression in the cancer sample compared to the normal adjacent). Table 2 shows overexpression of Lng129 in 38 lung cancer tissues compared with their respective normal adjacent (lung samples #2, 4, 6, 8, 9, 10, 11, 12, 13, 14, 17, 22, 24, 25, 26, 28, 30, 31, 33, 34, 35, 37, and 38). There is overexpression in the cancer tissue for 61% of the lung matching samples tested (23 out of total of 38 lung matching samples).


Altogether, the high level of tissue specificity, plus the mRNA differential expression in the lung matching samples tested are believed to make Lng129 a good marker for diagnosing, monitoring, staging, imaging and treating lung cancer.


Northern Analysis


Two transcripts ˜6.5 kb and ˜9 kb


DNA Sequence for Lng129


Sequence available from Incyte database.


Primers Used for QPCR Expression Analysis

Forward primerGCCTGTTTGGGAGATTAGATTTT(SEQ ID NO:24)Reverse primerGCCCAAACAGAACAGACTAAAAA(SEQ ID NO:25)ProbeAGGTTATTAGGTTATTATCTCTCTCTCCTGATTTTT(SEQ ID NO:26)CC


Example 3

Sequence 3


Lng112


Gene ID 983439

TABLE 1The absolute numbers are relative levels of expression ofLng112 in 12 normal different tissues. These RNA samples arecommercially available pools, originated by pooling samplesof a particular tissue from different individuals.TissueNORMALBrain0Heart0Kidney0Liver0Lung1.0Mammary0Muscle0Prostate0SmInt0Testis0Thymus0Uterus0
0 = negative


The relative levels of expression in Table 1 show that Lng112 mRNA expression is only detectable in lung compared with other normal tissues analyzed


The absolute numbers in Table 1 were obtained analyzing pools of samples of a particular tissue from different individuals. They can not be compared to the absolute numbers originated from RNA obtained from tissue samples of a single individual in Table 2.

TABLE 2The absolute numbers are relative levels of expression of Lng112in 49 pairs of matching samples. All the values are compared to normallung (calibrator). A matching pair is formed by mRNA from the cancersample for a particular tissue and mRNA from the normal adjacentsample for that same tissue from the same individual.MATCHINGNORMALSample IDCancer TypeTissueCANCERADJACENTLung 143LAdenocarcinomaLung 10.000.00Lung 60LAdenocarcinomaLung 213.009.00Lng AC82AdenocarcinomaLung 30.005.00Lng 60XLAdenocarcinomaLung 40.0019.00Lng AC66AdenocarcinomaLung 50.0059.00Lng AC69AdenocarcinomaLung 69.000.00Lng AC88AdenocarcinomaLung 716.0035.00Lng AC11AdenocarcinomaLung 80.0036.00Lng AC32AdenocarcinomaLung 90.0041.00Lng AC39AdenocarcinomaLung 100.005.00Lng AC94AdenocarcinomaLung 110.000.00Lng BA641Bronchio-alveolarLung 127.0014.00carcinomaLng SQ32Squamous cellLung 130.00228.00carcinomaLng SQ45Squamous cellLung 14368.002.00carcinomaLng SQ56Squamous cellLung 151.0053.00carcinomaLng SQ9XSquamous cellLung 160.002.00carcinomaLng SQ14Squamous cellLung 170.0021.00carcinomaLng SQ16Squamous cellLung 180.011.00carcinomaLng SQ80Squamous cellLung 196.007.00carcinomaLng C20XSquamous cellLung 200.000.00carcinomaLng 47XQSquamous cellLung 211.003.00carcinomaLng SQ44Squamous cellLung 220.000.00carcinomaLng SQ79Squamous cellLung 230.000.00carcinomaLng 90XSquamous cellLung 240.004.00carcinomaLng BR94Squamous cellLung 250.000.00carcinomaLng LC71Large cellLung 26178.004.00carcinomaLng LC80Large cellLung 270.000.00carcinomaLng LC109Large cellLung 281.0096.00carcinomaLung 77LLarge cellLung 290.000.00carcinomaLng 75XCMetastatic fromLung 300.0022.00bone cancerLng MT67Metastatic fromLung 311.0086.00renal cell cancerLng MT71Metastatic fromLung 320.0014.00melanomaBld 32XKBladder 10.000.00Cln AS45Colon 10.000.00Cvx KS52Cervix 10.000.00End 28XAEndometrium 10.000.00Kid 106XDKidney 10.000.00Liv 94XALiver 10.000.00Mam A06XMammary 10.000.00Ovr 103XOvary 10.000.00Pan 71XLPancreas 10.000.00Pan 77XPancreas 20.000.00Pro 20XBProstate 10.000.00Skn 287SSkin 10.000.00SmInt H89Sm. Int. 10.000.00Sto 531SStomach 10.000.00Thr 143NThyriod 10.000.00Tst 39XTestis 10.000.00Utr 135XOUterus 116.000.00
0 = Negative


In the analysis of matching samples, except 1 uterus cancer sample the only detection was in lung samples showing a high degree of tissue specificity for lung tissue. These results confirm the tissue specificity results obtained with normal pooled samples (Table 1).


Furthermore, we compared the level of mRNA expression in cancer samples and the isogenic normal adjacent tissue from the same individual. This comparison provides an indication of specificity for the cancer stage (e.g. higher levels of mRNA expression in the cancer sample compared to the normal adjacent). Table 2 shows overexpression of Lng112 in 4 lung cancer tissues compared with their respective normal adjacent tissue in 32 cancer matching pairs (lung samples # 2, 6, 14, and 26). There is overexpression in the cancer tissue for 12.5% of the lung matching samples tested (total of 32 lung matching samples).


Furthermore, we compared the level of mRNA expression in cancer samples and the isogenic normal adjacent tissue from the same individual. The result shows that Lng112 is expressed differentially in all 32 lung cancer tissues tested compared with their respective normal adjacent.


Altogether, the high level of tissue specificity, plus the mRNA differential expression in the lung matching samples tested are believed to make Lng112 a good marker for diagnosing, monitoring, staging, imaging and treating lung cancer.


Primers Used for QPCR Expression Analysis

Forward primerTGGTGGCGTTCCTCCTGTC(SEQ ID NO:27)Reverse primerCAGAGCCCTTCGTACTGGAACAC(SEQ ID NO:28)ProbeTCGTACAGGTCCTGGGTGCTCCACA(SEQ ID NO:29)


Example 4

Sequence 4


Lng114


Gene ID 236582

TABLE 1The absolute numbers are relative levels of expression ofLng114 in 12 normal different tissues. All the values arecompared to normal testis (calibrator). These RNA samplesare commercially available pools, originated by pooling samplesof a particular tissue from different individuals.TissueNORMALBrain0.09Heart0.00Kidney0.26Liver0.00Lung602.58Mammary0.35Muscle0.00Prostate0.00SmInt0.05Testis1.00Thyraus0.00Uterus1.27
0 = negative


The relative levels of expression in Table 1 show that Lng114 mRNA expression is highest in lung (602.58) compared with other normal tissues analyzed.


The absolute numbers in Table 1 were obtained analyzing pools of samples of a particular tissue from different individuals. They can not be compared to the absolute numbers originated from RNA obtained from tissue samples of a single individual in Table 2.

TABLE 2The absolute numbers are relative levels of expression of Lng114 in 78 pairsof matching samples, 1 normal ovary and 2 blood samples. All the values arecompared to normal testis (calibrator). A matching pair is formed by mRNAfrom the cancer sample for a particular tissue and mRNA from the normal adjacentsample for that same tissue from the same individual.MATCHINGNORMALSample IDCancer TypeTissueCANCERADJACENTNORMALLng 60LAdenocarcinomaLung 1121.5266.72Lng 143LAdenocarcinomaLung 2360.7925.99Lng 60XLAdenocarcinomaLung 379.81648.73Lng AC82AdenocarcinomaLung 437.53102.18Lng AC88AdenocarcinomaLung 5530.06992.55Lng AC66AdenocarcinomaLung 676.68257.93Lng AC69AdenocarcinomaLung 725.468.51Lng AC11AdenocarcinomaLung 854.19852.17Lng AC32AdenocarcinomaLung 9157.59193.34Lng AC94AdenocarcinomaLung 102272.40112.99Lng AC90AdenocarcinomaLung 11141.5338.85Lng AC39AdenocarcinomaLung 12198.098.40Lng 223LAdenocarcinomaLung 1310.6331.89Lng 528LAdenocarcinomaLung 14210.84274.37Lng BR26BronchogenicLung 150.00169.48carcinomaLng BA641Bronchio-alveolarLung 16316.2773.77carcinomaLng 315LSquamous cellLung 170.00469.51carcinomaLng SQ14Squamous cellLung 181016.9315.83carcinomaLng SQ56Squamous cellLung 190.78526.39carcinomaLng SQ9XSquamous cellLung 2052.8964.89carcinomaLng SQ80Squamous cellLung 2160.34962.07carcinomaLng SQ45Squamous cellLung 2297.01357.05carcinomaLng SQ16Squamous cellLung 2392.411833.01carcinomaLng SQ32Squamous cellLung 2423.7531.02carcinomaLng SQ79Squamous cellLung 2520.89142.52carcinomaLng 47XQSquamous cellLung 2642.52135.77carcinomaLng BR94Squamous cellLung 27211.50157.78carcinomaLng 90XSquamous cellLung 2880.7312.21carcinomaLng C20XSquamous cellLung 292.9915.24carcinomaLng SQ44Squamous cellLung 3094.030.00carcinomaLng SQ43Squamous cellLung 3127.1938.85carcinomaLng LC71Large cellLung 321217.752040.91carcinomaLng LC109Large cellLung 33160.424576.44carcinomaLng LC80Large cellLung 34955.43400.32carcinomaLng 77LLarge cellLung 3518.4478.52carcinomaLng 75XCMetastatic fromLung 36229.13398.93bone cancerLng MT67Metastatic fromLung 3769.071514.89renal cell cancerLng MT71Metastatic fromLung 3842.371393.99melanomaBld 46XKBladder 10.000.00Bld 66XBladder 20.000.00Blo B5Blood 10.00Blo B6Blood 20.00Cln AS43Colon 11.220.00Cln AS45Colon 20.000.00Cln AS46Colon 32.080.00Cln SG67Colon 41.491.39Cvx KS52Cervix 12.3911.47Cvx KS83Cervix 21.224.55Endo 28XAEndometrium 1108.382.86Endo 68XEndometrium 23.7312.64Kid10XDKidney 139.400.00Kid 109XDKidney 21.918.46Kid 107XDKidney 31.484.61Liv 15XALiver 10.030.07Liv 2O1LLiver 20.000.00Liv 174LLiver 30.000.00Mam 162XMammary 10.780.28Mam 173MMammary 21.000.00Mam 220Mammary 32.020.30Ovr 18GAOvary 10.00Ovr AO84Ovary 20.000.00Pro 101XBProstate 10.861.38Pro 109XBProstate 20.000.23Pro125XBProstate 30.000.08Pan 77XPancreas 10.000.00Skn 39ASkin 10.200.00Skn 39ABSkin 20.000.00Skn 248SSkin 30.000.00Smint 21XASm. Int. 10.1100.00Smint H89Sm. Int. 20.000.00Sto 264SStomach 11.045.98Sto 288SStomach 25.100.00Sto 115SStomach 35.030.75Thr 143NThyroid 10.000.94Thr 145TThyroid 21.892.50Thr 939TThyriod 31.520.00Tst 647TTestis 110.200.00Tst 39XTestis 28.200.00Tst 663TTestis 35.090.00Utr 141XOUterus 113.185.65Utr 135XOUterus 21.471.36
0 = Negative


In the analysis of matching samples, higher expression of lng114 is detected in lung samples showing a high degree of tissue specificity for lung tissue. These results confirm the tissue specificity results obtained with normal pooled samples (Table 1).


Furthermore, we compared the level of mRNA expression in cancer samples and the isogenic normal adjacent tissue from the same individual. This comparison provides an indication of specificity for the cancer stage (e.g. higher levels of mRNA expression in the cancer sample compared to the normal adjacent). Table 2 shows overexpression of Lng114 in 11 lung cancer tissues compared with their respective normal adjacent tissue in 35 cancer matching pairs (lung samples #1, 2, 7, 10, 11, 12, 15, 16, 26, 28, and 32). There is overexpression in the cancer tissue for 31% of the lung matching samples tested (total of 35 primary cancer lung matching samples).


Altogether, the high level of tissue specificity, plus the mRNA overexpression in 31% of the lung matching samples tested are believed to make Lng114 a good marker for diagnosing, monitoring, staging, imaging and treating lung cancer.


Primers Used for QPCR Expression Analysis

Forward primerCTTGGCAGCTCACATGGAAC(SEQ ID NO:30)Reverse primerCTGGGGTGTCTCTGTCACTCTC(SEQ ID NO:31)ProbeCCATGAAGTCCCACCCCTTTTCTCTG(SEQ ID NO:32)


Example 5

Sequence 5


Lng118


Gene ID 210995

TABLE 1The absolute numbers are relative levels of expression ofLng118 in 24 normal different tissues. These RNA samples arecommercially available pools, originated by pooling samplesof a particular tissue from different individuals.TissueNORMALAdrenal Gland0Bladder0Brain0.010Cervix0.010Colon0Endometrium0.010Esophagus0Heart0Kidney0.010Liver0Lung1.000Mammary Gland0.010Muscle0.0032Ovary0.005Pancreas0.005Prostate0.002Rectum0.004Small Intestine0Spleen0Stomach0.015Testis0.033Thymus0.001Trachea0.007Uterus0.005
0 = negative


The relative levels of expression in Table 1 show that Lng118 mRNA expression is high in lung compared with other normal tissues analyzed.


The absolute numbers in Table 1 were obtained analyzing pools of samples of a particular tissue from different individuals. They can not be compared to the absolute numbers originated from RNA obtained from tissue samples of a single individual in Table 2.

TABLE 2The absolute numbers are relative levels of expression of Lng118in 36 pairs of matching samples. All the values are compared to normallung (calibrator). A matching pair is formed by mRNA from the cancersample for a particular tissue and mRNA from the normal adjacentsample for that same tissue from the same individual.Sample IDCancer TypeTissueCANCERMATCHINGLng60LAdenocarcinomaLung 10.10.04LngAC66AdenocarcinomaLung 200.27LngAC69AdenocarcinomaLung 300.11Lng AC88AdenocarcinomaLung 40.050.13Lng 60XLAdenocarcinomaLung 500.08LngAC94AdenocarcinomaLung 600LngAC11AdenocarcinomaLung 700.02LngAC32AdenocarcinomaLung 800.05Lng 47XQAdenocarcinomaLung 900Lng223LAdenocarcinomaLung 1000.01Lng BR26Bronchio-alveolarLung 1100LngSQ45Squamous cellLung 120.540LngSQ16Squamous cellLung 1300LngSQ79Squamous cellLung 1400Lng LC71Large cell carcinomaLung 151.230.06Lng LC109Large cell carcinomaLung 1600.06Lng 75XCMetastatic fromLung 1700BldTR17Bladder 100Cvx KS52Cervix 100ClnSG45Colon 100Colon 200End 10479Endometrium 100Kid 106XDKidney 100.01Kid 5XDKidney 200Liv 187LLiver 100Liv175LLiver 200Mam S967Mammary 100Ovr A084Ovary 10.140Pan 71XLPancreas 100Pro 20XBProstate 10.020Pro 326Prostate 200SmInt H89Small00Sto 531SStomach 100Tst 39XTestis 100Thr 270TThyroid 10.020.01Thr 644T
0 = Negative


In the analysis of matching samples, higher expression of lng118 is detected in lung samples showing a high degree of tissue specificity for lung tissue. These results confirm the tissue specificity results obtained with normal pooled samples (Table 1).


Furthermore, we compared the level of mRNA expression in cancer samples and the isogenic normal adjacent tissue from the same individual. This comparison provides an indication of specificity for the cancer stage (e.g. higher levels of mRNA expression in the cancer sample compared to the normal adjacent). Table 2 shows differential expression of Lng118 in 17 lung cancer tissues compared with their respective normal adjacent tissue.


Altogether, the high level of tissue specificity, plus the mRNA differential expression in the lung matching samples tested are believed to make Lng118 a good marker for diagnosing, monitoring, staging, imaging and treating lung cancer.


DNA Sequence for Lng118


Sequence available from Incyte database.


Primers Used for QPCR Expression Analysis

Forward primerTGCAGCAGAAAGGGGAGAG(SEQ ID NO:33)Reverse primerTCCCCATTGCCCTCAAGT(SEQ ID NO:34)ProbeCGTGGGCACTCACCTCGGCACT(SEQ ID NO:35)


Example 6

Sequence 6


Lng121


Gene ID 208994

TABLE 1The absolute numbers are relative levels of expression ofLng121 in 24 normal different tissues. All the values arecompared to normal trachea (calibrator). These RNA samplesare commercially available pools, originated by pooling samplesof a particular tissue from different individuals.TissueNORMALAdrenal Gland0.01Bladder0.00Brain0.55Cervix0.09Colon0.02Endometrium1.74Esophagus0.08Heart0.00Kidney0.04Liver0.00Lung117.38Mammary Gland0.47Muscle0.36Ovary0.41Pancreas0.10Prostate0.93Rectum0.05Small Intestine0.09Spleen1.72Stomach0.12Testis3.24Thymus2.06Trachea1.00Uterus0.12
0 = negative


The relative levels of expression in Table 1 show that Lng121 mRNA expression is high in lung compared with most other normal tissues analyzed.


The absolute numbers in Table 1 were obtained analyzing pools of samples of a particular tissue from different individuals. They can not be compared to the absolute numbers originated from RNA obtained from tissue samples of a single individual in Table 2.

TABLE 2The absolute numbers are relative levels of expression of Lng121 in20 pairs of matching samples. All the values are compared to normaltrachea (calibrator). A matching pair is formed by mRNA from the cancersample for a particular tissue and mRNA from the normal adjacent samplefor that same tissue from the same individual.MATCHINGNORMALSample IDCancer TypeTissueCANCERADJACENTLng 60LAdenocarcinomaLung 19.9516.62Lng 143LAdenocarcinomaLung 21.075.41Lng 60XLAdenocarcinomaLung 312.775.08Lng AC88AdenocarcinomaLung 45.0631.89Lng AC66AdenocarcinomaLung 53.8522.32Lng AC32AdenocarcinomaLung 68.4687.12Lng 223LAdenocarcinomaLung 71.874.10Lng SQ14Squamous cellLung 82.9133.72carcinomaLng C20XSquamous cellLung 90.080.29carcinomaLng 77LLarge cell carcinomaLung 108.1316.35Lng LC71Large cell carcinomaLung 1147.843.69Lng 75XCMetastatic fromLung 123.4915.67melanomaCln AS43Colon 11.220.17Endo 12XAEndometrium 12.380.29Kid 107XDKidney 10.440.17Liv 187LLiver 10.031.06Mam 19DNMammary 11.410.58Ovr A084Ovary 10.760.28Pro 109XBProstate 10.190.27Tst 647TTestis 12.921.64
0 = Negative


In the analysis of matching samples, higher expression of lng121 is detected in lung samples showing a high degree of tissue specificity for lung tissue. These results confirm the tissue specificity results obtained with normal pooled samples (Table 1).


Furthermore, we compared the level of mRNA expression in cancer samples and the isogenic normal adjacent tissue from the same individual. This comparison provides an indication of specificity for the cancer stage (e.g. higher levels of mRNA expression in the cancer sample compared to the normal adjacent). Table 2 shows differential expression of Lng121 in 12 lung cancer tissues compared with their respective normal adjacent tissue.


Altogether, the high level of tissue specificity, plus the mRNA differential expression in the lung matching samples tested are believed to make Lng121 a good marker for diagnosing, monitoring, staging, imaging and treating lung cancer.


DNA sequence for Lng121


Sequence available from Incyte database.


Primers Used for QPCR Expression Analysis

Forward primerCAGGCTCATTTTATTGTGGTCAT(SEQ ID NO:36)Reverse primerCCCACACTGATTTAGGCACATAG(SEQ ID NO:37)ProbeTTTGAAGGAGGGCAGGAAAAACTATGTAAG(SEQ ID NO:38)


Example 7

Sequence 7


Lng124


Gene ID 1066498

TABLE 1The absolute numbers are relative levels of expression ofLng124 in 24 normal different tissues. All the values arecompared to normal lung (calibrator). These RNA samples arecommercially available pools, originated by pooling samplesof a particular tissue from different individuals.TissueNORMALAdrenal Gland0Bladder0Brain0Cervix0Colon0Endometrium0Esophagus0Heart0Kidney0Liver0Lung1.00Mammary Gland0Muscle0Ovary0Pancreas0Prostate0Rectum0Small Intestine0Spleen0Stomach0Testis0Thymus0Trachea0Uterus0
0 = negative


The relative levels of expression in Table 1 show that Lng124 mRNA expression is only detectable in lung compared with most other normal tissues analyzed.


The absolute numbers in Table 1 were obtained analyzing pools of samples of a particular tissue from different individuals. They can not be compared to the absolute numbers originated from RNA obtained from tissue samples of a single individual in Table 2.

TABLE 2The absolute numbers are relative levels of expression of Lng124in 40 pairs of matching samples. All the values are compared to normallung (calibrator). A matching pair is formed by mRNA from the cancersample for a particular tissue and mRNA from the normal adjacentsample for that same tissue from the same individual.MATCHINGNORMALSample IDCancer TypeTissueCANCERADJACENTLng60LAdenocarcinomaLung 10.640.62Lng 143LAdenocarcinomaLung 20.030.26LngAC66AdenocarcinomaLung 30.040.51Lng 60XLAdenocarcinomaLung 40.040.36Lng AC88AdenocarcinomaLung 50.111.05LngAC11AdenocarcinomaLung 60.122.48LngAC32AdenocarcinomaLung 70.220.64Lng 47XQAdenocarcinomaLung 80.190.13Lng AC39AdenocarcinomaLung 90.160.7Lng AC90AdenocarcinomaLung 100.10.12Lng223LAdenocarcinomaLung 110.050.17Lng SQ14Squamous cellLung 1200.69carcinomaLng SQ9XSquamous cellLung 130.150.12carcinomaLngSQ16Squamous cellLung 140.120.24carcinomaLngSQ79Squamous cellLung 150.10.42carcinomaLng SQ43Squamous cellLung 160.140.17carcinomaLng BR94Squamous cellLung 170.010.03carcinomaLng C20XSquamous cellLung 1800.02carcinomaLng LC109Large cellLung 190.060.74carcinomaBld 66XBladder 100Cvx NK23Cervix 100Cvx NK24Cervix 200ClnAS45Colon 200Cln RC24Colon 300End 8911Endometrium 100Kid 6XDKidney 100Kid 710KKidney 200Liv 94XALiver 100Mam 173MMammary 200Mam S123Mammary 300Ovr A082Ovary 100Ovr C179Ovary 200Ovr 130XOvary 30Pan 92XPancreas 100Pro 34BProstate 100Sto 531SStomach 100Sto AC93Stomach 200Sto 288SStomach 300Sto TA73Stomach 400Sto 288SStomach 500Sto 531SStomach 600Skn 287SSkin 100Thr692TThyroid 10.020
0 = Negative


In the analysis of matching samples, expression of lng124 is only detected in lung samples (except 1 thyroid cancer sample) showing a high degree of tissue specificity for lung tissue. These results confirm the tissue specificity results obtained with normal pooled samples (Table 1).


Furthermore, we compared the level of mRNA expression in cancer samples and the isogenic normal adjacent tissue from the same individual. This comparison provides an indication of specificity for the cancer stage (e.g. higher levels of mRNA expression in the cancer sample compared to the normal adjacent). Table 2 shows differential expression of Lng124 in all of the 19 lung cancer tissues compared with their respective normal adjacent tissue.


Altogether, the high level of tissue specificity, plus the mRNA differential expression in all of the lung matching samples tested are believed to make Lng124 a good marker for diagnosing, monitoring, staging, imaging and treating lung cancer.


Primers Used for QPCR Expression Analysis

Forward primerAGGGAGAGGAGCTATGGACGT(SEQ ID NO:39)Reverse primerTTTTGAGGCAAGACTCCATCTC(SEQ ID NO:40)ProbeCTGCCAAGGGAGAGAGTGAGGTAGGC(SEQ ID NO:41)


Example 8

Sequence 8


Lng126


Gene ID 287016

TABLE 1The absolute numbers are relative levels of expression ofLng126 in 24 normal different tissues. All the values arecompared to normal thymus (calibrator). These RNA samplesare commercially available pools, originated by pooling samplesof a particular tissue from different individuals.TissueNORMALAdrenal Gland11.92Bladder0.0Brain0.21Cervix0.7Colon0.06Endometrium6.36Esophagus0.04Heart0.06Kidney1.11Liver7.94Lung6.2Mammary Gland7.46Muscle0.78Ovary38.32Pancreas2.69Prostate5.21Rectum2.72Small0.6Spleen0.16Stomach0.93Testis3.2Thymus1.00Trachea4.61Uterus3.90
0 = negative


The relative levels of expression in Table 1 show that Lng126 mRNA expression is relatively high in lung, except adrenal gland and ovary, compared with other normal tissues analyzed.


The absolute numbers in Table 1 were obtained analyzing pools of samples of a particular tissue from different individuals. They can not be compared to the absolute numbers originated from RNA obtained from tissue samples of a single individual in Table 2.

TABLE 2The absolute numbers are relative levels of expressionof Lng126 in 20 pairs of matching samples. All the valuesare compared to normal thymus (calibrator). A matchingpair is formed by mRNA from the cancer sample for a particulartissue and mRNA from the normal adjacent sample for thatsame tissue from the same individual.MATCHINGNORMALSample IDCancer TypeTissueCANCERADJACENTLng 60LAdenocarcinomaLung 10.060.03Lng 143LAdenocarcinomaLung 27.340.45LngAC66AdenocarcinomaLung 30.100.07LngAC69AdenocarcinomaLung 40.330.04LngAC11AdenocarcinomaLung 50.720.30LngAC32AdenocarcinomaLung 60.140.10LngAC94AdenocarcinomaLung 70.110.01Lng223LAdenocarcinomaLung 80.010.01LngSQ45Squamous cellLung 90.430.16carcinomaLng SQ14Squamous cellLung 1011.352.61carcinomaLngSQ16Squamous cellLung 110.090.01carcinomaLngSQ79Squamous cellLung 1210.780.14carcinomaLng C20XSquamous cellLung 130.260.00carcinomaLng 77LLarge cellLung 141.327.14carcinomaBld 66XBladder 14.9243.56ClnAS45Colon 11.261.28Mam 19DNMammary 114.620.48Mam 220Mammary 20.330.61Mam S854Mammary 30.661.04
0 = Negative


In the analysis of matching samples, higher expression of lng126 is detected in lung samples showing a high degree of tissue specificity for lung tissue. These results confirm the tissue specificity results obtained with normal pooled samples (Table 1).


Furthermore, we compared the level of mRNA expression in cancer samples and the isogenic normal adjacent tissue from the same individual. This comparison provides an indication of specificity for the cancer stage (e.g. higher levels of mRNA expression in the cancer sample compared to the normal adjacent). Table 2 shows differential expression of Lng126 in 14 lung cancer tissues compared with their respective normal adjacent tissue.


Altogether, the high level of tissue specificity, plus the mRNA differential expression in the lung matching samples tested are believed to make Lng126 a good marker for diagnosing, monitoring, staging, imaging and treating lung cancer.


DNA Sequence for Lng126


Sequence available from Incyte database.


Primers Used for QPCR Expression Analysis

Forward primerTGGGGACAATATGGACCTCA(SEQ ID NO:42)Reverse primerGGCGAGTGTCTATGATGAACCT(SEQ ID NO:43)ProbeCAGGATCTGTGAGGATTTCATTTGGATACAT(SEQ ID NO:44)


Example 9

Sequence 9


Lng136


Gene ID 10717


ddx Lung Code SQLng001

TABLE 1The absolute numbers are relative levels of expression ofLng136 in 24 normal different tissues. All the values arecompared to normal spleen (calibrator). These RNA samplesare commercially available pools, originated by pooling samplesof a particular tissue from different individuals.TissueNORMALAdrenal Gland0.34Bladder0.03Brain0.66Cervix0.12Colon0.00Endometrium0.08Esophagus0.05Heart0.02Kidney0.01Liver0.00Lung8.54Mammary1.32Muscle0.00Ovary0.07Pancreas0.86Prostate0.15Rectum0.02Small Int.0.05Spleen1.0Stomach0.77Testis1.22Thymus0.19Trachea0.16Uterus0.03
0 = negative


The relative levels of expression in Table 1 show that Lng136 mRNA expression is high in lung compared with most other normal tissues analyzed.


The absolute numbers in Table 1 were obtained analyzing pools of samples of a particular tissue from different individuals. They can not be compared to the absolute numbers originated from RNA obtained from tissue samples of a single individual in Table 2.

TABLE 2The absolute numbers are relative levels of expression of Lng136in 60 pairs of matching samples. All the values are compared to normalspleen (calibrator). A matching pair is formed by mRNA from the cancersample for a particular tissue and mRNA from the normal adjacentsample for that same tissue from the same individual.MATCHINGNORMALSample IDCancer TypeTissueCANCERADJACENTLng60LAdenocarcinomaLung 15.924.77Lng 143LAdenocarcinomaLung 22.202.58Lng AC82AdenocarcinomaLung 32.626.68LngAC66AdenocarcinomaLung 41.363.61Lng 60XLAdenocarcinomaLung 50.664Lng AC88AdenocarcinomaLung 63.6115.94LngAC69AdenocarcinomaLung 77.497.19LngAC11AdenocarcinomaLung 81.3131.45LngAC32AdenocarcinomaLung 97.419.51Lng AC90AdenocarcinomaLung 1016.345.26Lng223LAdenocarcinomaLung 112.411.49LngAC94AdenocarcinomaLung 121.472.09Lng BR26Bronchio-alveolarLung 1306.5carcinomaLngSQ45Squamous cellLung 1419.972.18carcinomaLng SQ14Squamous cellLung 151.7613.04carcinomaLng SQ56Squamous cellLung 162.1812.73carcinomaLngSQ16Squamous cellLung 170.545.30carcinomaLng SQ32Squamous cellLung 183.3114.17carcinomaLng AC39Squamous cellLung 193.4315.08carcinomaLng 47XQSquamous cellLung 200.745.3carcinomaLngSQ79Squamous cellLung 212.538.49carcinomaLng C20XSquamous cellLung 220.070.22carcinomaLng SQ44Squamous cellLung 231.483.59carcinomaLng SQ43Squamous cellLung 241.450.91carcinomaLng LC71Large cellLung 2511.7910.67carcinomaLng 77LLarge cellLung 269.253.11carcinomaLng LC109Large cellLung 276.8736.89carcinomaLng MT67Metastatic fromLung 282.935.01renal cell cancerLng MT71Metastatic fromLung 290.191.23melanomaBldTR14Bladder 10.250.94Cvx NK24Cervix 10.460.14Cvx KS52Cervix20.070.03ClnAS43Colon10.020.07ClnAS45Colon20.100.06ClnAS46Colon30.160.13ClnAS67Colon40.040.14ClnAS89Colon50.100.36End 8911Endometrium 10.060.18End 28XAEndometrium 20.230.12Kid 5XDKidney10.011.41Kid 109XDKidney20.470.39Liv15XALiver10.110.03Liv 174LLiver 20.010.01Mam S123Mammary 10.190.17Mam 162XMammary 20.150.17Ovr C179Ovary 100Ovr 130XOvary 20.580Pan 71 XLPancreas 10.070Pan 92XPancreas 26.941.62Pro 326Prostate 10.040.12Pro 109XBProstate 20.010.01Skn 278SSkin 10.040.1SmInt H89Small intestine 10.130.04SmInt 21XASmall intestine 20.180Sto TA73Stomach 12.94.18Sto 758SStomach 20.771.53Tst647TTestis 10.580.38Tst 39XTestis 20.501.02Thr 270TThyroid 10.030.02Utr135XOUterus 10.20.48
0 = Negative


In the analysis of matching samples, higher expression of lng136 is detected in lung samples showing a high degree of tissue specificity for lung tissue. These results confirm the tissue specificity results obtained with normal pooled samples (Table 1).


Furthermore, we compared the level of mRNA expression in cancer samples and the isogenic normal adjacent tissue from the same individual. This comparison provides an indication of specificity for the cancer stage (e.g. higher levels of mRNA expression in the cancer sample compared to the normal adjacent). Table 2 shows differential expression of Lng136in 29 lung cancer tissues compared with their respective normal adjacent tissue.


Altogether, the high level of tissue specificity, plus the mRNA differential expression in the lung matching samples tested are believed to make Lng136 a good marker for diagnosing, monitoring, staging, imaging and treating lung cancer


DNA Sequence for Lng136


Sequence available from Incyte database.


Primers Used for QPCR Expression Analysis

Forward primerCTCCGTGGCTCGTGCTT(SEQ ID NO:45)Reverse primerCGCTTTCTTTTTGCCCTCTTGT(SEQ ID NO:46)


Example 10

Sequence 10


Lng143


Gene ID 24945


ddx Lung Code SQLng006

TABLE 1The absolute numbers are relative levels of expression ofLng143 in 24 normal different tissues. All the values arecompared to normal pancreas (calibrator). These RNA samplesare commercially available pools, originated by pooling samplesof a particular tissue from different individuals.TissueNORMALAdrenal Gland0.83Bladder0.04Brain1.11Cervix0.20Colon0.01Endometrium2.49Esophagus0.01Heart0.09Kidney0.34Liver0.23Lung6.15Mammary Gland2.34Muscle0.44Ovary4.20Pancreas1.00Prostate6.34Rectum1.14Small Intestine0.16Spleen6.63Stomach1.13Testis3.12Thymus7.39Trachea2.77Uterus6.04
0 = negative


The relative levels of expression in Table 1 show that Lng143 mRNA expression is much higher in lung compared with most other normal tissues analyzed.


The absolute numbers in Table 1 were obtained analyzing pools of samples of a particular tissue from different individuals. They can not be compared to the absolute numbers originated from RNA obtained from tissue

TABLE 2The absolute numbers are relative levels of expression of Lng143 in 78 pairsof matching samples, 2 blood samples and 2 normal ovary samples. All the valuesare compared to normal pancreas (calibrator). A matching pair is formed bymRNA from the cancer sample for a particular tissue and mRNA from the normaladjacent sample for that same tissue from the same individual.MATCHINGNORMALSample IDCancer TypeTissueCANCERADJACENTNORMALLng 60LAdenocarcinomaLung 10.540.41Lng 143AdenocarcinomaLung 20.410.08Lng 60XLAdenocarcinomaLung 31.090.86Lng AC82AdenocarcinomaLung 43.250.09Lng AC88AdenocarcinomaLung 53.990.93Lng AC66AdenocarcinomaLung 60.990.42Lng AC69AdenocarcinomaLung 72.360.50Lng AC11AdenocarcinomaLung 82.671.80Lng AC32AdenocarcinomaLung 93.020.43Lng AC39AdenocarcinomaLung 109.350.13Lng AC94AdenocarcinomaLung 110.580.26Lng AC90AdenocarcinomaLung 123.850.01Lng 223LAdenocarcinomaLung 130.320.02Lng 528LAdenocarcinomaLung 1410.523.77Lng BR26BronchogenicLung 158.400.28carcinomaLng BA641Bronchio-alveolarLung 162.580.37carcinomaLng SQ45Squamous cellLung 174.611.53carcinomaLng 315LSquamous cellLung 181.151.16carcinomaLng SQ14Squamous cellLung 191.830.78carcinomaLng SQ9XSquamous cellLung 202.700.14carcinomaLng SQ56Squamous cellLung 212.501.53carcinomaLng SQ80Squamous cellLung 222.690.77carcinomaLng SQ32Squamous cellLung 237.701.51carcinomaLng SQ16Squamous cellLung 240.700.04carcinomaLng SQ79Squamous cellLung 253.610.92carcinomaLng 90XSquamous cellLung 261.240.23carcinomaLng 47XQSquamous cellLung 271.900.13carcinomaLng BR94Squamous cellLung 282.870.00carcinomaLng C20XSquamous cellLung 290.050.04carcinomaLng SQ44Squamous cellLung 300.212.13carcinomaLng SQ43Squamous cellLung 312.860.04carcinomaLng LC71Large cellLung 321.941.82carcinomaLng LC109Large cellLung 334.044.30carcinomaLng LC80Large cellLung 346.130.51carcinomaLng 77LLarge cellLung 350.031.08carcinomaLng 75XCMetastatic fromLung 360.150.19bone cancerLng MT71Metastatic fromLung 375.960.74renal cell cancerLng MT67Metastatic fromLung 3812.301.18melanomaBld46XKBladder 10.030.02BldTR14Bladder 22.891.51Blo B5Blood 121.19Blo B6Blood 241.21Cvx KS52Cervix 15.781.44Cvx KS83Cervix 217.754.29ClnAS43Colon 13.420.10ClnAS45Colon 20.170.13ClnAS46Colon 32.291.92ClnAS67Colon 40.200.33ClnAS89Colon 50.080.12End 10479Endometrium 125.634.63End 28XAEndometrium 26.252.46End 68XEndometrium 36.4311.24Kid10XDKidney 13.731.07Kid 109XDKidney 22.904.82Liv15XALiver 10.190.08liv 174 LLiver 20.990.76Mam 173 MMammary 10.760.47Mam 220Mammary 20.110.23Mam 355Mammary 31.080.19Mam 976MMammary 40.020.16ovr 180Bovary 116.11Ovr 18GAOvary 215.14Ovr A084Ovary 38.065.58Pan 77XPancreas 13.942.51Pan 92XPancreas 24.981.70Pro 101XBProstate 13.692.15Pro 109XBProstate 20.160.23Pro 125XBProstate 30.110.12Pro 13XBProstate 40.040.31Skn 39ASkin 11.190.08Skn 816SSkin 20.950.01SmInt 21XASmall Intestine 10.750.10SmInt H89Small Intestine 20.660.46Sto 115SStomach 11.911.20Sto 264SStomach 20.740.99Sto288SStomach 32.780.06Tst647TTestis 11.872.68Tst 663TTestis 27.890.66Thr 270TThyroid 12.012.13Thr 939TThyroid 20.500.55Utr135XOUterus 13.526.06Utr 141XOUterus 22.592.57
0 = Negative


In the analysis of matching samples, higher expression of lng143 is detected in lung samples showing a high degree of tissue specificity for lung tissue. These results confirm the tissue specificity results obtained with normal pooled samples (Table 1).


Furthermore, we compared the level of mRNA expression in cancer samples and the isogenic normal adjacent tissue from the same individual. This comparison provides an indication of specificity for the cancer stage (e.g. higher levels of mRNA expression in the cancer sample compared to the normal adjacent). Table 2 shows differential expression of Lng143 in 38 lung cancer tissues compared with their respective normal adjacent tissue.


Altogether, the high level of tissue specificity, plus the mRNA differential expression in the lung matching samples tested are believed to make Lng143 a good marker for diagnosing, monitoring, staging, imaging and treating lung cancer.


DNA Sequence for Lng143


Sequence available from Incyte database.


Primers Used for QPCR Expression Analysis

Forward primerCCGACCTTGAGATTATTCCTGT(SEQ ID NO:47)Reverse primerGCACCACTTAAACCAAATCCA(SEQ ID NO:48)ProbeTGCTGCCAACACCACTTCTCCATCT(SEQ ID NO:49)


Example 11

Sequence 11


Lng144


Gene ID 52017


ddx Lung Code SQlng007

TABLE 1The absolute numbers are relative levels of expression ofLng144 in 24 normal different tissues. All the values arecompared to normal uterus (calibrator). These RNA samplesare commercially available pools, originated by pooling samplesof a particular tissue from different individuals.TissueNORMALAdrenal Gland0.04Bladder1.29Brain0.44Cervix0.85Colon0.00Endometrium0.43Esophagus0.05Heart0.06Kidney0.18Liver0.30Lung1.35Mammary Gland1.04Muscle0.34Ovary0.29Pancreas0.77Prostate0.93Rectum0.26Small0.11Spleen3.92Stomach0.30Testis1.1Thymus0.93Trachea0.69Uterus1.00
0 = negative


The relative levels of expression in Table 1 show that Lng144 mRNA expression is high in lung compared with other normal tissues analyzed.


The absolute numbers in Table 1 were obtained analyzing pools of samples of a particular tissue from different individuals. They can not be compared to the absolute numbers originated from RNA obtained from tissue samples of a single individual in Table 2.

TABLE 2The absolute numbers are relative levels of expression of Lng144in 30 pairs of matching samples. All the values are compared to normaluterus (calibrator). A matching pair is formed by mRNA from the cancersample for a particular tissue and mRNA from the normal adjacent samplefor that same tissue from the same individual.MATCHINGSampleNORMALIDCancer TypeTissueCANCERADJACENTLng 60LAdenocarcinomaLung 10.650.30Lng 143LAdenocarcinomaLung 20.290.17Lng AC66AdenocarcinomaLung 300.34Lng AC69AdenocarcinomaLung 41.411.89Lng AC11AdenocarcinomaLung 52.823.11Lng AC32AdenocarcinomaLung 61.091.27Lng AC94AdenocarcinomaLung 72.200.84Lng 223LAdenocarcinomaLung 80.300.27Lng BR26Bronchio-alveolarLung 91.250.36carcinomaLng SQ45Squamous cellLung 102.961.26carcinomaLng SQ9XSquamous cellLung 111.490.30carcinomaLng SQ80Squamous cellLung 121.881.51carcinomaLng SQ16Squamous cellLung 130.480.47carcinomaLng SQ79Squamous cellLung 142.770.00carcinomaLng 90XSquamous cellLung 150.090.29carcinomaLng SQ43Squamous cellLung 160.810.26carcinomaLng LC71Large cellLung 170.981.65Carcinoma ILng LC109Large cellLung 180.381.63carcinoma IIIALng MT71MetastaticLung 190.130.20from melanomaLng MT67MetastaticLung 200.630.77from renalcancerBld46XKBladder 10.240.08BldTR14Bladder 20.180.95ClnAS45Colon20.120.05ClnAS46Colon30.210.98ClnAS67Colon40.090.18ClnAS89Colon 50.383.31ClnAS43Colon50.180.47Liv15XALiver 10.470.05Tst647TTestis 12.100.26Utr135XOUterus 10.810.80
0 = Negative


In the analysis of matching samples, higher expression of lng144 is detected in lung samples showing a high degree of tissue specificity for lung tissue. These results confirm the tissue specificity results obtained with normal pooled samples (Table 1).


Furthermore, we compared the level of mRNA expression in cancer samples and the isogenic normal adjacent tissue from the same individual. This comparison provides an indication of specificity for the cancer stage (e.g. higher levels of mRNA expression in the cancer sample compared to the normal adjacent). Table 2 shows differential expression of Lng144 in 20 lung cancer tissues compared with their respective normal adjacent tissue.


Altogether, the high level of tissue specificity, plus the mRNA differential expression in the lung matching samples tested are believed to make Lng144 a good marker for diagnosing, monitoring, staging, imaging and treating lung cancer.


DNA Sequence for Lng144


Sequence available from Incyte database.


Primers Used for QPCR Expression Analysis

Forward primerTGCTGCCACAAACCGAGA(SEQ ID NO:50)Reverse primerTTGGGAGGGTTGGTTGGTT(SEQ ID NO:51)ProbeTTTTGAGGGCACTAGGGAACGATCTGT(SEQ ID NO:52)


Example 12

Sequence 12


Lng138


Gene ID 460254


ddx Lung Code SQlng110

TABLE 1The absolute numbers are relative levels of expression ofLng138 in 24 normal different tissues. All the values arecompared to normal spleen (calibrator). These RNA samplesare commercially available pools, originated by pooling samplesof a particular tissue from different individuals.TissueNORMALAdrenal Gland0.00Bladder0.00Brain0.04Cervix0.09Colon0.00Endometrium0.31Esophagus0.00Heart0.00Kidney0.03Liver0.00Lung1.35Mammary Gland0.02Muscle0.03Ovary1.15Pancreas0.03Prostate0.27Rectum0.02Small Intestine0.02Spleen1.00Stomach0.05Testis0.08Thymus1.00Trachea0.12Uterus0.24
0 = negative


The relative levels of expression in Table 1 show that Lng138 mRNA expression is high in lung compared with other normal tissues analyzed.


The absolute numbers in Table 1 were obtained analyzing pools of samples of a particular tissue from different individuals. They can not be compared to the absolute numbers originated from RNA obtained from tissue samples of a single individual in Table 2.

TABLE 2The absolute numbers are relative levels of expression of Lng138in 50 pairs of matching samples. All the values are compared to normalspleen (calibrator). A matching pair is formed by mRNA from the cancersample for a particular tissue and mRNA from the normal adjacentsample for that same tissue from the same individual.MATCHINGNORMALSample IDCancer TypeTissueCANCERADJACENTLng 60LAdenocarcinomaLung 11.660.82Lng 143LAdenocarcinomaLung 20.060.03Lng AC82AdenocarcinomaLung 30.020.03Lng AC66AdenocarcinomaLung 40.10.79Lng AC69AdenocarcinomaLung 50.560.65Lng 60XLAdenocarcinomaLung 60.020.15Lng AC94AdenocarcinomaLung 70.670.65Lng AC11AdenocarcinomaLung 80.413.32Lng AC32AdenocarcinomaLung 90.471.57Lng 47XQAdenocarcinomaLung 100.030.26Lng 223LAdenocarcinomaLung 110.30.16Lng BR26BronchogenicLung 120.120.26carcinomaLng BA641Bronchio-alveolarLung 130.170.1carcinomaLngSQ45Squamous cellLung 145.30.22carcinomaLng SQ14Squamous cellLung 150.020.4carcinomaLng SQ9XSquamous cellLung 160.380.06carcinomaLng SQ56Squamous cellLung 170.050.73carcinomaLng SQ16Squamous cellLung 180.040.4carcinomaLng SQ32Squamous cellLung 190.110.68carcinomaLng SQ80Squamous cellLung 200.170.38carcinomaLng SQ79Squamous cellLung 210.290.79carcinomaLng SQ43Squamous cellLung 220.190.01carcinomaLng BR94Squamous cellLung 230.10carcinomaLng 90XSquamous cellLung 240.010.02carcinomaLng LC71Large cellLung 250.20.47carcinomaLng LC109Large cellLung 260.090.54carcinomaLng MT67MetastaticLung 270.40.11from renalcarcinomaBld46XKBladder 100BldTR14Bladder 20.10.04ClnAS43Colon 10.110.06ClnAS45Colon 20.030.03ClnAS46Colon 30.080.04ClnAS67Colon 40.050.11ClnAS89Colon 50.020.08End 28XAEndometrium 10.080.12Kid 109XDKidney 10.120.05Kid 10XDKidney 20.090.02Liv15XALiver 10.010Mam 173MMammary 10.030.02Mam 220Mammary 20.010.26Mam 355Mammary 30.10.03Ovr A084Ovary 10.110.08Pro 101XBProstate 10.070.09Pro 109XBProstate 20.010.01Pro 125XBProstate 300.01Sto 115SStomach 10.070.07Sto 246SStomach 20.160.1Sto 288SStomach 30.070.01Tst 647TTestis 20.130.02Utr135XOUterus 10.410.18
0 = Negative


In the analysis of matching samples, higher expression of lng138 is detected in lung samples showing a high degree of tissue specificity for lung tissue. These results confirm the tissue specificity results obtained with normal pooled samples (Table 1).


Furthermore, we compared the level of mRNA expression in cancer samples and the isogenic normal adjacent tissue from the same individual. This comparison provides an indication of specificity for the cancer stage (e.g. higher levels of mRNA expression in the cancer sample compared to the normal adjacent). Table 2 shows differential expression of Lng138 in 27 lung cancer tissues compared with their respective normal adjacent tissue.


Altogether, the high level of tissue specificity, plus the mRNA differential expression in the lung matching samples tested are believed to make Lng138 a good marker for diagnosing, monitoring, staging, imaging and treating lung cancer.


DNA Sequence for Lng138


Sequence available from Incyte database.


Primers Used for QPCR Expression Analysis

Forward primerCCTGATACCTTTAACCAATGCTCT(SEQ ID NO:53)Reverse primerTTGGGTAGTATCAAATGGGTAAGG(SEQ ID NO:54)ProbeCCTGTCCTTCTCCTTTGGCTTATGCTATCC(SEQ ID NO:55)


Example 13

Sequence 13


Lng137


Gene ID 179090


ddx Lung Code SQLng012

TABLE 1The absolute numbers are relative levels of expression ofLng137 in 24 normal different tissues. All the values arecompared to normal spleen (calibrator). These RNA samplesare commercially available pools, originated by pooling samplesof a particular tissue from different individuals.TissueNORMALAdrenal Gland0.042Bladder0.063Brain0.285Cervix0.196Colon0.080Endometrium0.956Esophagus0.025Heart0.010Kidney0.046Liver0.035Lung0.204Mammary Gland0.142Muscle0.092Ovary0.760Pancreas0.084Prostate0.355Rectum0.357Small Intestine0.074Spleen1.000Stomach0.103Testis2.612Thymus10.853Trachea0.076Uterus0.235
0 = negative


The relative levels of expression in Table 1 show that Lng137 mRNA expression is relatively high in lung compared with most other normal tissues analyzed.


The absolute numbers in Table 1 were obtained analyzing pools of samples of a particular tissue from different individuals. They can not be compared to the absolute numbers originated from RNA obtained from tissue samples of a single individual in Table 2.

TABLE 2The absolute numbers are relative levels of expression of Lng137in 70 pairs of matching samples. All the values are compared to normalspleen (calibrator). A matching pair is formed by mRNA from the cancersample for a particular tissue and mRNA from the normal adjacent samplefor that same tissue from the same individual.Sample IDCancer TypeTissueCANCERMATCHINGLng60LAdenocarcinomaLung 10.920.67Lng 143LAdenocarcinomaLung 20.530.04Lng AC82AdenocarcinomaLung 32.70LngAC66AdenocarcinomaLung 41.620.34LngAC69AdenocarcinomaLung 53.180.79Lng AC88AdenocarcinomaLung 61.870.32Lng 60XLAdenocarcinomaLung 73.420.24LngAC94AdenocarcinomaLung 800.21LngAC11AdenocarcinomaLung 923.432.76LngAC32AdenocarcinomaLung 105.170.63Lng 47XQAdenocarcinomaLung 112.030Lng AC90AdenocarcinomaLung 124.690Lng AC39AdenocarcinomaLung 132.480.22Lng223LAdenocarcinomaLung 141.890Lng 528LAdenocarcinomaLung 151.470Lng BR26Bronchio-alveolarLung 1613.180.47Lng BA641Squamous cellLung 170.970.18Lng 315LSquamous cellLung 180.630.62LngSQ45Squamous cellLung 193.218.7Lng SQ14Squamous cellLung 202.280.29Lng SQ9XSquamous cellLung 2110.06Lng SQ56Squamous cellLung 2210.270.67LngSQ16Squamous cellLung 232.060Lng SQ32Squamous cellLung 241.340.58Lng SQ80Squamous cellLung 255.210.29LngSQ79Squamous cellLung 2610.590.93Lng C20XSquamous cellLung 2700Lng SQ43Squamous cellLung 2816.560.04Lng BR94Squamous cellLung 299.920Lng SQ44Squamous cellLung 300.136.23Lng 90XSquamous cellLung 310.310Lng 77LLarge cellLung 320.071.75carcinomaLng LC71Large cellLung 331.930.55carcinomaLng LC109Large cellLung 3413.980.79carcinomaLng LC80Large cellLung 357.310carcinomaLng MT67MetastaticLung 362.030.17from boneLng MT71MetastaticLung 371.550from renalLng 75XCMetastaticLung 380.040.17fromBld46XKBladder 100BldTR14Bladder 24.472.79Cvx KS52Cervix 140.53Cvx KS83Cervix 26.430.92ClnAS43Colon 112.550.77ClnAS45Colon 21.510.35ClnAS46Colon 3181.6ClnAS67Colon 41.65.48ClnAS89Colon 50.780.04End 28XAEndometrium3.660.64End 8911Endometrium1.682.36Kid 109XDKidney 10.520.44Kid 10XDKidney 20.510.07Liv15XALiver 100.02Liv174LLiver 20.160.09Mam 162XMammary 10.030.1Mam 355Mammary 20.130.08Ovr A084Ovary 11.421.3Pan 71XLPancreas 11.010.38Pan 92XPancreas 20.641.16Pro 109XBProstate 10.080.15Pro 125XBProstate 20.030.03SmIntSmall0.330.03SmInt H89Small2.940.12Sto 758SStomach 12.890.13Sto 288SStomach 21.030.05Tst 47TTestis 13.930.56Tst 39XTestis 21.91.39Thr 143NThyroid 10.160.36Thr 270TThyroid 20.350Utr 35XOUterus 12.92.62Utr 141XOUterus 20.870.38
0 = Negative


In the analysis of matching samples, higher expression of lng137 are detected in lung samples showing a high degree of tissue specificity for lung tissue. These results confirm the tissue specificity results obtained with normal pooled samples (Table 1).


Furthermore, we compared the level of mRNA expression in cancer samples and the isogenic normal adjacent tissue from the same individual. This comparison provides an indication of specificity for the cancer stage (e.g. higher levels of mRNA expression in the cancer sample compared to the normal adjacent). Table 2 shows overexpression of Lng137 in 31 lung cancer tissues compared with their respective normal adjacent tissue in 38 cancer matching pairs (lung samples #2, 3, 4, 5, 6, 7, 9, 10, 11, 12, 13, 14, 15, 16, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 31, 33, 34, 35, 36, and 37). There is overexpression in the cancer tissue for 82% of the lung matching samples tested (total of 38 lung matching samples).


Altogether, the high level of tissue specificity, plus the mRNA overexpression in 82% of the lung matching samples tested are believed to make Lng137 a good marker for diagnosing, monitoring, staging, imaging and treating lung cancer.


Northern Analysis


One transcript ˜3.4 kb


DNA Sequence for Lng137


Sequence available from Incyte database.


Primers Used for QPCR Expression Analysis

Forward primerCTCGGATATGATTAAAGAGTTTCG(SEQ ID NO:56)Reverse primerTCCACTGTGCTGTTTGTTGTT(SEQ ID NO:57)ProbeATTGGCGTGCTCTTTGTAACTCTGAGA(SEQ ID NO:58)


Example 14

Sequence 14


Lng142


Gene ID 6348


ddx Lung Code SQlng004

TABLE 1The absolute numbers are relative levels of expression ofLng142 in 24 normal different tissues. All the values arecompared to normal lung (calibrator). These RNA samples arecommercially available pools, originated by pooling samplesof a particular tissue from different individuals. TissueNORMALAdrenal Gland0.02Bladder0.00Brain0.00Cervix0.00Colon0.00Endometrium0.00Esophagus0.00Heart0.00Kidney0.00Liver0.00Lung1.00Mammary Gland0.01Muscle0.00Ovary0.03Pancreas0.01Prostate0.02Rectum0.00Small Intestine0.01Spleen0.00Stomach0.00Testis0.03Thymus0.00Trachea0.01Uterus0.03
0 = negative


The relative levels of expression in Table 1 show that Lng142 mRNA expression is high in lung compared with most other normal tissues analyzed.


The absolute numbers in Table 1 were obtained analyzing pools of samples of a particular tissue from different individuals. They can not be compared to the absolute numbers originated from RNA obtained from tissue samples of a single individual in Table 2.

TABLE 2The absolute numbers are relative levels of expression of Lng142in 20 pairs of matching samples. All the values are compared to normallung (calibrator). A matching pair is formed by mRNA from the cancersample for a particular tissue and mRNA from the normal adjacentsample for that same tissue from the same individual. MATCHINGNORMALSample IDCancer TypeTissueCANCERADJACENTLng 60LAdenocarcinomaLung 10.380.22Lng AC66AdenocarcinomaLung 20.000.05Lng AC69AdenocarcinomaLung 30.000.00Lng AC11AdenocarcinomaLung 40.001.37Lng AC32AdenocarcinomaLung 50.050.16Lng AC94AdenocarcinomaLung 60.080.00Lng 223LAdenocarcinomaLung 70.030.00Lng SQ45Squamous cellLung 83.820.02carcinomaLng SQ16Squamous cellLung 90.000.00carcinomaLng SQ79Squamous cellLung 100.000.11carcinomaBld 46XKBladder 10.000.00Bld TR14Bladder 20.000.00Cln AS45Colon 10.000.00Cln AS46Colon 20.000.00Cln AS67Colon 30.000.01Cln AS89Colon 40.010.02Cln AS43Colon 50.000.00Liv 15XALiver 10.000.00Tst 647TTestis 10.000.05Utr 135XOUterus 10.170.00
0 = Negative


In the analysis of matching samples, higher expression of lng142 is detected in lung samples showing a high degree of tissue specificity for lung tissue. These results confirm the tissue specificity results obtained with normal pooled samples (Table 1).


Furthermore, we compared the level of mRNA expression in cancer samples and the isogenic normal adjacent tissue from the same individual. This comparison provides an indication of specificity for the cancer stage (e.g. higher levels of mRNA expression in the cancer sample compared to the normal adjacent). Table 2 shows differential expression of Lng142 in 10 lung cancer tissues compared with their respective normal adjacent tissue.


Altogether, the high level of tissue specificity, plus the mRNA differential expression in the lung matching samples tested are believed to make Lng142 a good marker for diagnosing, monitoring, staging, imaging and treating lung cancer.


DNA Sequence for Lng142


Sequence available from Incyte database.


Primers Used for QPCR Expression Analysis

Forward primerTGGCTAAAATAGGTCTTGTAGGGA(SEQ ID NO:59)Reverse primerCAAGGAGGGGGCATTTGTA(SEQ ID NO:60)ProbeTCCTTTCCTTGGCAATCTCCTCTCCTG(SEQ ID NO:61)


Example 15

Sequence 15


Lng140


Gene ID 94694


ddx Lung Code SQLng005

TABLE 1The absolute numbers are relative levels of expression of Lng140in 24 normal different tissues. All the values are comparedto normal mammary gland (calibrator). These RNA samples arecommercially available pools, originated by pooling samplesof a particular tissue from different individuals.TissueNORMALAdrenal Gland0Bladder0.00Brain0.00Cervix0.00Colon0.00Endometrium0.14Esophagus0.00Heart0.00Kidney0.00Liver0.00Lung183.55Mammary Gland1.00Muscle0.00Ovary0.00Pancreas0.00Prostate0.10Rectum0.06Small0.03Spleen0.00Stomach0.02Testis0.01Thymus0.00Trachea3.72Uterus0.00
0 = negative


The relative levels of expression in Table 1 show that Lng140 mRNA expression is much higher in lung compared with most other normal tissues analyzed.


The absolute numbers in Table 1 were obtained analyzing pools of samples of a particular tissue from different individuals. They can not be compared to the absolute numbers originated from RNA obtained from tissue samples of a single individual in Table 2.

TABLE 2The absolute numbers are relative levels of expression of Lng140 in 78 pairs ofmatching samples, 2 blood samples, 1 normal ovary and 1 cancer ovary sample. Allthe values are compared to normal mammary gland (calibrator). A matching pair isformed by mRNA from the cancer sample for a particular tissue and mRNA from thenormal adjacent sample for that same tissue from the same individual.MATCHINGNORMALSample IDCancer TypeTissueCANCERADJACENTNORMALLng 60LAdenocarcinomaLung 121.563.54Lng 143LAdenocarcinomaLung 20.007.31Lng 60XLAdenocarcinomaLung 388.0319.84Lng AC82AdenocarcinomaLung 44.61122.36Lng AC88AdenocarcinomaLung 54.6170.77Lng AC66AdenocarcinomaLung 638.197.46Lng AC69AdenocarcinomaLung 736.0025.99Lng AC11AdenocarcinomaLung 81287.18280.14Lng AC32AdenocarcinomaLung 9208.660.00Lng AC94AdenocarcinomaLung 1039.81153.28Lng AC90AdenocarcinomaLung 1178.25420.22Lng AC39AdenocarcinomaLung 12600.495.17Lng 223LAdenocarcinomaLung 135.603.56Lng 528LAdenocarcinomaLung 146.1728.05Lng BR26BronchogenicLung 154.6823.18carcinomaLng BA641Bronchio-alveolarLung 16263.204.11carcinomaLng 315LSquamous cellLung 170.003.77carcinomaLng SQ14Squamous cellLung 180.745.60carcinomaLng SQ56Squamous cellLung 1936.25186.75carcinomaLng SQ9XSquamous cellLung 2077.981.99carcinomaLng SQ80Squamous cellLung 2120.3235.02carcinomaLng SQ45Squamous cellLung 22153.2880.73carcinomaLng SQ16Squamous cellLung 239.4513.04carcinomaLng SQ32Squamous cellLung 243213.6699.04carcinomaLng SQ79Squamous cellLung 25594.2848.17carcinomaLng 47XQSquamous cellLung 2647.840.00carcinomaLng BR94Squamous cellLung 274.660.00carcinomaLng 90XSquamous cellLung 280.006.41carcinomaLng C20XSquamous cellLung 292.350.00carcinomaLng SQ44Squamous cellLung 306.591.55carcinomaLng SQ43Squamous cellLung 3125.190.00carcinomaLng LC71Large cellLung 321408.5597.01carcinomaLng LC109Large cellLung 3385.92922.88carcinomaLng LC80Large cellLung 3499.3911.16carcinomaLng 77LLarge cellLung 358.6911.35carcinomaLng 75XCMetastatic fromLung 360.000.00bone cancerLng MT67Metastatic fromLung 370.002.28renal cell cancerLng MT71Metastatic fromLung 381.560.00melanomaBld46XKBladder 10.000.00BldTR14Bladder 20.00168.90Blo B5Blood 10.00Blo B6Blood 20.00Cvx KS52Cervix 185.330.00Cvx KS83Cervix 223.510.00ClnAS43Colon1259.570.00ClnAS45Colon20.000.00ClnAS46Colon314.520.00ClnAS67Colon45.9041.64ClnAS89Colon55.136.54End 10479Endometrium 10.000.00End 28XAEndometrium 238.451.29End 68XEndometrium 30.002.30Kid10XDKidney 10.000.00Kid 109XDKidney 20.000.00Liv15XALiver 10.000.00liv 174Liver 20.000.00LMam 173MMammary 10.870.00Mam 220Mammary 20.000.00Mam 355Mammary 30.000.00Mam 976MMammary 40.000.00ovr 180Bovary 10.000.00Ovr 18GAOvary 20.00Ovr A084Ovary 336.50Pan 77XPancreas 10.000.00Pan 92XPancreas 246.530.00Pro 101XBProstate 10.291.43Pro 109XBProstate 20.000.00Pro 125XBProstate 31.301.97Pro 13XBProstate 40.000.00Skn 39ASkin 10.000.00Skn 816SSkin 20.000.00SmInt 21XASmall Intestine 11.451.70SmInt H89Small Intestine 279.071.17Sto 115SStomach 1109.1410.16Sto 264SStomach 22.530.00Sto288SStomach 30.000.00Tst647TTestis 10.000.00Tst 663TTestis 20.000.00Thr 270TThyroid 10.000.00Thr 939TThyroid 20.000.00Utr135XOUterus 13.890.00Utr 141XOUterus 20.291.43
0 = Negative


In the analysis of matching samples, the higher expression level of lng140 is detected in lung samples showing a high degree of tissue specificity for lung tissue. These results confirm the tissue specificity results obtained with normal pooled samples (Table 1).


Furthermore, we compared the level of mRNA expression in cancer samples and the isogenic normal adjacent tissue from the same individual. This comparison provides an indication of specificity for the cancer stage (e.g. higher levels of mRNA expression in the cancer sample compared to the normal adjacent). Table 2 shows overexpression of Lng140 in 20 lung cancer tissues compared with their respective normal adjacent tissue in 38 cancer matching pairs (lung samples #1, 3, 6, 8, 9, 12, 13, 16, 20, 22, 24-15 27, 29-32, 34, and 38). There is overexpression in the cancer tissue for 5.3% of the lung matching samples tested (total of 38 lung matching samples).


Altogether, the high level of tissue specificity, plus the mRNA overexpression in 53% of the lung matching samples tested are believed to make Lng140a good marker for diagnosing, monitoring, staging, imaging and treating lung cancer.


Primers Used for QPCR Expression Analysis

Forward primerCCTCTGAAGAAACGATCACAACA(SEQ ID NO:62)Reverse primerATTCCAGCCTGAGTCACACAGA(SEQ ID NO:63)ProbeACCAAGGAGAAACAAAACCAAGCAGCA(SEQ ID NO:64)


Example 16

Sequence 16


Lng151


Gene ID 145812


ddx Lung Code SQlng008

TABLE 1The absolute numbers are relative levels of expression ofLng151 in 24 normal different tissues. All the values arecompared to normal thymus (calibrator). These RNA samplesare commercially available pools, originated by pooling samplesof a particular tissue from different individuals.TissueNORMALAdrenal Gland0.01Bladder0.01Brain0.03Cervix0.07Colon0.01Endometrium0.16Esophagus0.02Heart0.00Kidney0.01Liver0.00Lung0.17Mammary Gland0.06Muscle0.04Ovary0.44Pancreas0.05Prostate0.04Rectum0.03Small Intestine0.01Spleen0.13Stomach0.02Testis0.03Thymus1.00Trachea0.09Uterus0.09
0 = negative


The relative levels of expression in Table 1 show that Lng15l mRNA expression is high in lung compared with most other normal tissues analyzed.


The absolute numbers in Table 1 were obtained analyzing pools of samples of a particular tissue from different individuals. They can not be compared to the absolute numbers originated from RNA obtained from tissue samples of a single individual in Table 2.

TABLE 2The absolute numbers are relative levels of expressionof Lng151 in 20 pairs of matching samples. All the valuesare compared to normal thymus (calibrator). A matchingpair is formed by mRNA from the cancer sample for a particulartissue and mRNA from the normal adjacent sample for thatsame tissue from the same individual.MATCHINGNORMALSample IDCancer TypeTissueCANCERADJACENTLng 60LAdenocarcinomaLung 10.350.18Lng AC66AdenocarcinomaLung 20.370.34Lng AC69AdenocarcinomaLung 31.990.32Lng AC11AdenocarcinomaLung 41.131.11Lng AC32AdenocarcinomaLung 50.750.23Lng AC94AdenocarcinomaLung 60.20.1Lng 223LAdenocarcinomaLung 70.060Lng SQ45Squamous cellLung 82.450.94carcinomaLng SQ16Squamous cellLung 90.180.05carcinomaLng SQ79Squamous cellLung 101.230.62carcinomaBld 46XKBladder 10.060Bld TR14Bladder 20.270.37Cln AS43Colon 50.270.04Cln AS45Colon 10.020.04Cln AS46Colon 20.040.15Cln AS67Colon 30.030.28Cln AS89Colon 40.050.32Liv 15XALiver 10.220.08Tst 647TTestis 10.260.21Utr 135X0Uterus 11.141.06
0 = Negative


In the analysis of matching samples, higher expression of lng15l is detected in lung samples showing a high degree of tissue specificity for lung tissue. These results confirm the tissue specificity results obtained with normal pooled samples (Table 1).


Furthermore, we compared the level of mRNA expression in cancer samples and the isogenic normal adjacent tissue from the same individual. This comparison provides an indication of specificity for the cancer stage. (e.g. higher levels of mRNA expression in the cancer sample compared to the normal adjacent). Table 2 shows differential expression of Lng151 in 10 lung cancer tissues compared with their respective normal adjacent tissue.


Altogether, the high level of tissue specificity, plus the mRNA differential expression in the lung matching samples tested are believed to make Lng151 a good marker for diagnosing, monitoring, staging, imaging and treating lung cancer.


DNA Sequence for Lng151


Sequence available from Incyte database.


Primers Used for QPCR Expression Analysis

Forward primerTGAGGAGAAAGAAGGGAATCAC(SEQ ID NO:65)Reverse primerTCCTAAGGTAGCACTATTTGGAGAC(SEQ ID NO:66)ProbeAGCAATGAAGAATGAACTTGGAGTAAAGAGTCA(SEQ ID NO:67)


Example 17

Sequence 17


Lng150


Gene ID 10713


ddx Lung Code SQlng002

TABLE 1The absolute numbers are relative levels of expression ofLng150 in 24 normal different tissues. All the values arecompared to normal testis (calibrator). These RNA samplesare commercially available pools, originated by pooling samplesof a particular tissue from different individuals.TissueNORMALAdrenal Gland0.00Bladder0.04Brain0.01Cervix0.00Colon0.00Endometrium0.04Esophagus0.00Heart0.00Kidney0.01Liver0.00Lung0.01Mammary Gland0.00Muscle0.00Ovary0.00Pancreas0.00Prostate0.03Rectum0.00Small Intestine0.00Spleen0.00Stomach0.00Testis1.00Thymus0.00Trachea0.01Uterus0.07
0 = negative


The relative levels of expression in Table 1 show that Lng150 mRNA expression is detected in lung and is not detectable in adrenal gland, cervix, colon, esophagus, heart, liver, mammary gland, muscle, ovary, pancreas, rectum, small intestine, spleen, stomach, and thymus.


The absolute numbers in Table 1 were obtained analyzing pools of samples of a particular tissue from different individuals. They can not be compared to the absolute numbers originated from RNA obtained from tissue samples of a single individual in Table 2.

TABLE 2The absolute numbers are relative levels of expression of Lng150in 40 pairs of matching samples. All the values are compared to normaltestis (calibrator). A matching pair is formed by mRNA from the cancersample for a particular tissue and mRNA from the normal adjacent samplefor that same tissue from the same individual.MATCHINGNORMALSample IDCancer TypeTissueCANCERADJACENTLng 60LAdenocarcinomaLung 10.000.01Lng AC88AdenocarcinomaLung 20.020.00Lng AC66AdenocarcinomaLung 30.000.24Lng AC69AdenocarcinomaLung 40.020.00LngAC11AdenocarcinomaLung 50.000.20Lng AC32AdenocarcinomaLung 60.040.00Lng AC39AdenocarcinomaLung 70.000.00Lng AC94AdenocarcinomaLung 80.000.00Lng AC90AdenocarcinomaLung 90.000.00Lng 223LAdenocarcinomaLung 100.000.00Lng BR26Bronchio-alveolarLung 110.020.02carcinomaLng SQ45Broncho-genicLung 121.080.04carcinomaLng SQ9XSquamous cellLung 130.000.00carcinomaLng SQ80Squamous cellLung 140.030.00carcinomaLng SQ16Squamous cellLung 150.480.00carcinomaLng SQ79Squamous cellLung 160.000.00carcinomaLng 47XQSquamous cellLung 170.000.00carcinomaLng SQ43Squamous cellLung 180.000.00carcinomaBld 46XKBladder 10.000.00Bld TR14Bladder 20.000.54Blad66XBladder 30.0000.00ClnAS43Colon 134.130.00Cln AS45Colon 20.000.00Cln AS46Colon 30.040.00Cln AS67Colon 42.270.01Cln AS89Colon 50.151.61Cln DC63Colon 60.020.00Endo 68XEndometrium10.110.22Endo 12XAEndometrium20.030.06Kid6XDKidney10.010.03Kid710KKidney20.000.00Liv 15XALiver 10.060.01Liv201LLiver20.000.03Mam986Mammary10.000.00Sto 288SStomach10.000.00Sto531SStomach20.000.02Tst39XTestis10.030.07Tst 647TTestis 20.020.09Thr590DThyroid 10.010.00Utr135XOUterus 20.001.25
0 = Negative


In the analysis of matching samples, higher expression of lng150 is detected in lung samples showing a relatively high degree of tissue specificity for lung tissue. These results confirm the tissue specificity results obtained with normal pooled samples (Table 1).


Furthermore, we compared the level of mRNA expression in cancer samples and the isogenic normal adjacent tissue from the same individual. This comparison provides an indication of specificity for the cancer stage (e.g. higher levels of mRNA expression in the cancer sample compared to the normal adjacent). Table 2 shows differential expression of Lng150 in 18 lung cancer tissues compared with their respective normal adjacent tissue.


Altogether, the high level of tissue specificity, plus the mRNA differential expression in the lung matching samples tested are believed to make Lng150 a good marker for diagnosing, monitoring, staging, imaging and treating lung cancer.


DNA Sequence for Lng150


Primers Used for QPCR Expression Analysis

Forward primerATGGGCAGGTCTTTCTTTCC(SEQ ID NO:68)Reverse primerAGGCAGTTCTGTTACCCCACTA(SEQ ID NO:69)ProbeTGTGCTAAGGACAGGATTGGTTGGGTA(SEQ ID NO:70)


Example 18

Sequence 18


Lng141


Gene ID 20152


ddx Lung Code SQlng003

TABLE 1The absolute numbers are relative levels of expression ofLng141 in 24 normal different tissues. All the values arecompared to normal brain (calibrator). These RNA samples arecommercially available pools, originated by pooling samplesof a particular tissue from different individuals.TissueNORMALAdrenal Gland0.04Bladder0.00Brain1.00Cervix0.77Colon0.02Endometrium0.36Esophagus0.00Heart0.02Kidney0.05Liver0.00Lung3.45Mammary Gland0.99Muscle0.31Ovary2.23Pancreas0.06Prostate0.31Rectum0.65Small Intestine0.04Spleen0.70Stomach0.07Testis0.28Thymus0.91Trachea0.69Uterus1.27
0 = negative


The relative levels of expression in Table 1 show that Lng14l mRNA expression is high in lung compared with most other normal tissues analyzed.


The absolute numbers in Table 1 were obtained analyzing pools of samples of a particular tissue from different individuals. They can not be compared to the absolute numbers originated from RNA obtained from tissue samples of a single individual in Table 2.

TABLE 2The absolute numbers are relative levels of expression of Lng141in 50 pairs of matching samples. All the values are compared to normalbrain (calibrator). A matching pair is formed by mRNA from the cancersample for a particular tissue and mRNA from the normal adjacentsample for that same tissue from the same individual.MATCHINGNORMALSample IDCancer TypeTissueCANCERADJACENTLng 60LAdenocarcinomaLung 17.141.89Lng 143LAdenocarcinomaLung 20.110.18Lng 60XLAdenocarcinomaLung 30.190.39Lng AC82AdenocarcinomaLung 40.290.00Lng AC66AdenocarcinomaLung 55.061.62Lng AC69AdenocarcinomaLung 627.004.17Lng AC11AdenocarcinomaLung 71.523.43Lng AC32AdenocarcinomaLung 82.142.94Lng AC94AdenocarcinomaLung 92.454.35Lng 223LAdenocarcinomaLung 100.001.21Lng BR26Bronchio-alveolarLung 111.150.00carcinomaLng BA641BronchogenicLung 1219.840.80carcinomaLng SQ45Squamous cellLung 1331.027.78carcinomaLng SQ14Squamous cellLung 140.290.58carcinomaLng SQ9XSquamous cellLung 150.540.50carcinomaLng SQ56Squamous cellLung 161.080.52carcinomaLng SQ80Squamous cellLung 170.550.85carcinomaLng SQ32Squamous cellLung 180.680.91carcinomaLng SQ16Squamous cellLung 190.900.79carcinomaLng SQ79Squamous cellLung 208.116.87carcinomaLng 90XSquamous cellLung 210.000.38carcinomaLng 47XQSquamous cellLung 220.240.28carcinomaLng BR94Squamous cellLung 230.300.00carcinomaLng SQ43Squamous cellLung 242.860.19carcinomaLng LC71Large cellLung 250.622.30carcinomaLng LC109Large cellLung 260.091.33carcinomaLng MT67Metastasis fromLung 270.630.67renal carcinomaBld TR14Bladder 20.000.00Bld 46XKBladder 30.000.00Cln AS89Colon 12.4011.55Cln AS67Colon 21.431.89Cln AS45Colon 30.000.27Cln AS46Colon 40.000.00Cln AS43Colon 56.250.00End 28XAEndometrium11.251.59kid 10XDKidney10.281.02Kid 109XDKidney20.230.64Liv 15XALiver 11.220.84Mam173MMammary 10.190.47Mam 220Mammary 20.000.31Mam 355Mammay 30.360.09Ovr A084Ovary 13.220.94Pro 101XBProstate 10.5558.28Pro 109 XBProstate 20.110.21Pro 125XBProstate 30.240.26Sto 115SStomach 10.300.26Sto 264SStomach 20.350.26Sto 288SStomach 30.060.00Tst 647TTestis 16.873.96Utr 135XOUterus 10.006.02
0 = Negative


In the analysis of matching samples, higher expression of lng141 is detected in lung samples showing a relatively high degree of tissue specificity for lung tissue. These results confirm the tissue specificity results obtained with normal pooled samples (Table 1).


Furthermore, we compared the level of mRNA expression in cancer samples and the isogenic normal adjacent tissue from the same individual. This comparison provides an indication of specificity for the cancer stage (e.g. higher levels of mRNA expression in the cancer sample compared to the normal adjacent). Table 2 shows differential expression of Lng141 in 27 lung cancer tissues compared with their respective normal adjacent tissue.


Altogether, the high level of tissue specificity, plus the mRNA differential expression in the lung matching samples tested are believed to make Lng141 a good marker for diagnosing, monitoring, staging, imaging and treating lung cancer.


DNA Sequence for Lng141


Sequence available from Incyte database.


Primers Used for QPCR Expression Analysis

Forward primerACTGCCCACCACGCTTTATA(SEQ ID NO:71)Reverse primerTGAGGGTGGGGAGAGGTTAC(SEQ ID NO:72)ProbeAGTCACATTATTAGAGGTTCGCATCTCAGG(SEQ ID NO:73)


Claims
  • 1. A LSG comprising: (a) a polynucleotide of SEQ ID NO:1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 74, or a variant thereof; (b) a polypeptide expressed by a polynucleotide of SEQ ID NO:1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or 74, or a variant thereof; or (c) a polynucleotide which is capable of hybridizing under stringent conditions to the antisense sequence of SEQ ID NO: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 74.
  • 2. The LSG of claim 1 wherein the polypeptide comprises SEQ ID NO: 75, 76, 77, 78, 79, 80, 81, 82, 83, or 84.
  • 3. A method for diagnosing the presence of lung cancer in a patient comprising: (a) determining levels of a LSG of claim 1 in cells, tissues or bodily fluids in a patient; and (b) comparing the determined levels of LSG with levels of LSG in cells, tissues or bodily fluids from a normal human control, wherein a change in determined levels of LSG in said patient versus normal human control is associated with the presence of lung cancer.
  • 4. A method of diagnosing metastases of lung cancer in a patient comprising: (a) identifying a patient having lung cancer that is not known to have metastasized; (b) determining levels of a LSG of claim 1 in a sample of cells, tissues, or bodily fluid from said patient; and (c) comparing the determined LSG levels with levels of LSG in cells, tissue, or bodily fluid of a normal human control, wherein an increase in determined LSG levels in the patient versus the normal human control is associated with a cancer which has metastasized.
  • 5. A method of staging lung cancer in a patient having lung cancer comprising: (a) identifying a patient having lung cancer; (b) determining levels of a LSG of claim 1 in a sample of cells, tissue, or bodily fluid from said patient; and (c) comparing determined LSG levels with levels of LSG in cells, tissues, or bodily fluid of a normal human control, wherein an increase in determined LSG levels in said patient versus the normal human control is associated with a cancer which is progressing and a decrease in the determined LSG levels is associated with a cancer which is regressing or in remission.
  • 6. A method of monitoring lung cancer in a patient for the onset of metastasis comprising: (a) identifying a patient having lung cancer that is not known to have metastasized; (b) periodically determining levels of a LSG of claim 1 in samples of cells, tissues, or bodily fluid from said patient; and (c) comparing the periodically determined LSG levels with levels of LSG in cells, tissues, or bodily fluid of a normal human control, wherein an increase in any one of the periodically determined LSG levels in the patient versus the normal human control is associated with a cancer which has metastasized.
  • 7. A method of monitoring a change in stage of lung cancer in a patient comprising: (a) identifying a patient having lung cancer; (b) periodically determining levels of a LSG of claim 1 in cells, tissues, or bodily fluid from said patient; and (c) comparing the periodically determined LSG levels with levels of LSG in cells, tissues, or bodily fluid of a normal human control, wherein an increase in any one of the periodically determined LSG levels in the patient versus the normal human control is associated with a cancer which is progressing in stage and a decrease is associated with a cancer which is regressing in stage or in remission.
  • 8. A method of identifying potential therapeutic agents for use in imaging and treating lung cancer comprising screening compounds for an ability to bind to or decrease expression of a LSG of claim 1 relative to the LSG in the absence of the compound wherein the ability of the compound to bind to the LSG or decrease expression of the LSG is indicative of the compound being useful in imaging and treating lung cancer.
  • 9. An antibody which specifically binds a polypeptide encoded by a LSG of claim 1.
  • 10. The antibody of claim 9 wherein the polypeptide comprises SEQ ID NO: 75, 76, 77, 78, 79, 80, 81, 82, 83 or 84.
  • 11. A method of imaging lung cancer in a patient comprising administering to the patient an antibody of claim 9.
  • 12. The method of claim 11 wherein said antibody is labeled with paramagnetic ions or a radioisotope.
  • 13. A method of treating lung cancer in a patient comprising administering to the patient a compound which downregulates expression or activity of a LSG of claim 1.
  • 14. A method of inducing an immune response against a target cell expressing a LSG of claim 1 comprising delivering to a human patient an immunogenically stimulatory amount of a LSG polypeptide so that an immune response is mounted against the target cell.
  • 15. The method of claim 14 wherein the LSG polypeptide comprises SEQ ID NO:75, 76, 77, 78, 79, 80, 81, 82, 83 or 84.
  • 16. A vaccine for treating lung cancer comprising a LSG of claim 1.
Parent Case Info

This application claims the benefit of priority from U.S. Provisional Application Ser. No. 60/228,378, filed Aug. 28, 2000, which is herein incorporated in its entirety.

Provisional Applications (1)
Number Date Country
60228378 Aug 2000 US
Continuations (1)
Number Date Country
Parent 09940227 Aug 2001 US
Child 10933058 Sep 2004 US