ANTISENSE OLIGONUCLEOIDES OF GLUTATHIONE S-TRANSFERASES FOR CANCER TREATMENT

Information

  • Patent Application
  • 20220073931
  • Publication Number
    20220073931
  • Date Filed
    January 30, 2020
    5 years ago
  • Date Published
    March 10, 2022
    3 years ago
Abstract
The present invention relates to the identification of glutathione S transferase in tumors containing the same to be treated to inhibit protein expression of GSTs proteins and induce cell death and decrease tumor volume.
Description
FIELD OF THE INVENTION

The present invention is based on the identification of glutathione S transferase (GSTs) in carcinogenic tumors and a novel treatment for mammals, which inhibits the protein expression of GSTs proteins. The treatment is carried out through the use of antisense oligonucleotides directed to the messenger ribonucleic acids (mRNA) of the GSTM3 and GSTP1, leading to a reduced proliferation of cancer cells and a decrease in tumor progression. In addition, this reduction in proliferation extends to cancers resistant to conventional therapies.


BACKGROUND OF THE INVENTION

According to the World Health Organization (WHO), cancer is a generic term that designates a broad group of diseases that can affect any part of the body; they are also called malignant tumors or malignancies. A feature that defines cancer is an altered cell division extending beyond its usual limits, being able to invade adjacent parts of the body or spread to other organs, a process called metastasis. Metastases are the leading cause of death from cancer.


The WHO considers that the most common cancers are lung, breast, colorectal, prostate, stomach, liver, esophagus, cervical, thyroid, bladder, non-Hodgkin lymphoma, pancreas, leukemia, kidney, uterine body, oropharynx, cerebral and central nervous system, ovarian, melanoma, gallbladder, larynx, multiple myeloma, nasopharyngeal, laryngopharynx, Hodgkin lymphoma, testicles, salivary glands, vulva, Kaposi sarcoma, penis, mesothelioma, and vaginal.


Therefore, there is a need for drugs and treatments for mammals that have even been diagnosed with cancer.


As an example, one of the most common treatments for cervical cancer includes a chemoradiation therapy based on cisplatin. In addition, this treatment is regularly the only option to treat cervical cancer in advanced stages, and in most cases the disease cannot be eradicated (see Green, J. A., Kirwan, J. M., Tierney, J. F., Symonds, P., Fresco, L., Collingwood, M., & Williams, C. J. (2001). Survival and recurrence after concomitant chemotherapy and radiotherapy for cancer of the uterine cervix: a systematic review and meta-analysis. Lancet, 358(9284), 781-786.


There is a vast literature and patent application documents related to the treatment of cancers, for example, U.S. patent application Ser. No. 15/270,774 of ZHI-MING ZHENG et al., Refers to polynucleotide markers that can be detected two and can be used for the diagnosis of pre-cancers associated with the Human Papillomavirus and cancers associated with the Human Papillomavirus, such as cervical cancer and cervical intraepithelial malignancies as well as methods of treating such cancers. However, said document, like others, does not indicate the treatment of cancers in mammals already diagnosed with them.


Mexican patent application No. MX/a/2014/004285 refers to a method for the diagnosis of cervical cancer comprising performing an electrophoretic polyacrylamide gel run of a serum sample of a subject suspected of having cervical cancer, as well as determining the presence of at least one protein of molecular weight selected from the group of 60 kDa and 50 kDa. This document is intended to diagnose cervical cancer early, but not to treat mammals that have already been diagnosed with cervical cancer.


European Patent No. EP1531843 refers to the hematological study in oncoginecology, which can be used in patients with cervical cancer recurrence to evaluate the effectiveness of anti-tumor treatment and predict the course of the tumor process. The method makes it possible to carry out the selection of the most important rational method of antitumor impact and minimize the development of a number of side effects. Said document does not describe selecting candidates for said treatment to improve its effectiveness.


Patent document No. JP2005189228 provides a method and kit for diagnosing cancers including non-small cell lung cancer, esophageal cancer, laryngeal cancer, pharyngeal cancer, lingual cancer, stomach cancer, kidney cancer, large intestine cancer, cervical cancer, brain tumor, pancreatic cancer and bladder cancer, which are provided with an immunological technique that uses monoclonal antibodies against AKR1B10. Said document claims a protein consisting of the amino acid sequence of SEQ ID NO:1 in a biological sample, or a protein consisting of an amino acid sequence in which one or more amino acids are deleted, substituted or added in the amino acid sequence of SEQ ID NO:1 and has an activity of aldocete lactase. The protein is one that has the amino acid sequence shown in SEQ ID NO:1, which is detected by an immunological method. The document refers to diagnosing the types of cancer mentioned there, but not treating a mammal that has already been diagnosed with cancer.


GSTs are a family of enzymes that exhibit various functions, including the detoxification of xenobiotic compounds, evasion of the immune system and inhibition of apoptosis. In various types of cancer, it has been reported that members of the glutathione S-transferase (GST) family are overexpressed and in most cases they are related to a poor prognosis and resistance to chemotherapy (see Cabelguenne et al., 2001; Huang, Tan, Thiyagarajan, & Bay, 2003; Meding et al., 2012; Pectasides, Kamposioras, Papaxoinis, & Pectasides, 2008). In particular, it has been reported that GSTP1 and GSTM3 are deregulated in cancer cells, such as: triple negative breast cancer, prostate cancer, lung cancer and colorectal cancer (see Loktionov, a, Watson, M. a, Gunter, M., Stebbings, W. S., Speakman, C. T., & Bingham, S. a. (2001) Glutathione-S-transferase gene polymorphisms in colorectal cancer patients: interaction between GSTM1 and GSTM3 allele variants as a risk-modulating factor Carcinogenesis, 22(7), 1053-1060.


In addition, it is known that the GSTP1 protein plays a regulatory role through interaction with the TRAF2 protein and decreased signal transduction of the receptors in the TNF-α and JNK pathways, which are responsible for the activation of the apoptosis (see Adler, V., Yin, Z., Fuchs, S. Y., Benezra, M., Rosario, L., Tew, K. D., Ronai, Z. (1999). Regulation of JNK signaling by GSTp. The EMBO journal, 18(5), 1321-1334). On the other hand, it has been observed that the overexpression of GSTM3 in colon cancer is considered a marker of regional lymph node metastases (see Meding, S., Balluff, B., Elsner, M., Schöne, C., Rauser, S., Nitsche, U., . . . Walch, A. (2012). Tissue-based proteomics reveals FXYD3, S100A11 and GSTM3 as novel markers for regional lymph node metastasis in colon cancer. Journal of Pathology, 228(4), 459-470), and the subexpression of GSTM3 in urinary bladder cancer is associated with longer survival (see Mitra, A. P., Pagliarulo, V., Yang, D., Waldman, F. M., Datar, R. H., Skinner, D. G., . . . Cote, R. J. (2009). Generation of a concise gene panel for outcome prediction in urinary bladder cancer. Journal of Clinical Oncology, 27(24), 3929-3937).


Recently antisense molecules capable of inhibiting gene expression with great specificity have been used and, due to this, many research efforts related to the modulation of gene expression by antisense oligonucleotides (OAS) are being made. Some of these OAS focus on the inhibition of specific genes such as oncogenes or viral genes. Antisense oligonucleotides are directed against RNA (sense chain) or against DNA, wherein they form triple structures that inhibit transcription by RNA polymerase II. To achieve a desired effect on the negative regulation of the specific gene, oligonucleotides should promote the decomposition of the targeted mRNA or block the translation of that mRNA, thus avoiding de novo synthesis of the unwanted target protein (see US 20120029060 A1)


International publication No. WO2017091885, which describes the use of antisense oligonucleotides, provides compounds, compositions and methods for modulating the expression of the monocarboxylate transporter 4 (MCT4). In particular, this invention relates to antisense oligonucleotides (OAS) capable of modulating the expression of human MCT4 mRNA and uses and methods thereof for the treatment of various indications, including various cancers. In particular, the invention relates to therapies and treatment methods for cancers such as prostate cancer, including castration-resistant prostate cancer (CRPC).


Protein overexpression of GSTM3 and GSTP1 proteins during tumor progression (hereinafter identified as PT) play a regulatory role through interaction with proteins, for example TRAF2/6 proteins and, therefore, a evasion of signal transduction of apoptosis activation, favoring cell survival and PT (Wu Y, Fan Y, Xue B, Luo L, Shen J, Zhang S, Jiang Y, Yin Z. Human glutathione S-transferase P1-1 interacts with TRAF2 and regulates TRAF2-ASK1 signals. Oncogene. 2006; 25: 5787-800. Doi: 10.1038/sj.onc.1209576, Although GSTM3 interacts with TRAF6). In addition, GST's expression is involved in the modulation of detoxification processes in cancer cells and, therefore, participates in the chemoresistance response of conventional therapies in the cancer patients.


SUMMARY OF THE INVENTION

The present invention relates to the identification of GSTM3 and/or GSTP1 proteins in cancers, to provide a treatment with antisense oligonucleotides directed to said proteins (GSMT3 and GSTP1) to mammals identified as candidates for said treatment. At least one or more of said combined oligonucleotides block a protein specifically or both proteins.


According to a first aspect, the invention is directed to GSTM3 and GSTP1 proteins to be used as therapeutic targets and/or prognostic factors for mammals with cancer.


In the present invention, xenografted cancer cell lines in immunosuppressed mice were used to analyze, through their proteomics, the differences in protein expression during tumor progression. In this analysis it was found that glutathione S transferase P1 and M3 (hereafter referred to as GSTP1 and GSTM3, respectively) were some of the proteins that consistently increased their levels during tumor growth. In addition, it was found that through “inhibition of genes” (knock-down) of said proteins, they play a critical role for cell survival and tumor progression. Also, the abundance of the levels of these proteins in cancer biopsies was correlated with the survival of the patients. Therefore, it was shown that GSTP1 and GSTM3 proteins are also useful for prognostic purposes and that they are excellent candidates for target gene therapy for cancer.


In a second aspect, the present invention provides the use of antisense oligonucleotides of glutathione S transferases, preferably GSTM3 and GSTP1, without being limited thereto, for the treatment of cancers, in candidate subjects who have previously been diagnosed with cancer, wherein antisense oligonucleotides are between 15-50 nucleotides in length.


In another aspect, the present invention provides a method of treatment for cancer comprising: a) the extraction of the protein from the tumor tissue, b) carrying out an analysis by immunodetection techniques such as, for example, spotting of bands by western (Western blot), immunohistochemistry, ELISA, etc., to identify if the tumor has GSTM3 and/or GSTP1 proteins and c) administer the antisense oligonucleotides for said proteins.


In another aspect, the present invention provides a kit for identifying a candidate subject to be treated with the oligonucleotides of the present invention comprising at least one antisense oligonucleotide of the glutathione S transferases, preferably GSTM3 and GSTP1, without being limited thereto., a protein extraction solution, at least two antibodies for the identification of GSTM3 and GSTP1 proteins and optionally a secondary antibody, and a colorimetric developing solution for western (western blot) or immunohistochemistry (IHQ) staining.


Additional aspects and advantages of the present invention will be better understood by persons skilled in the art in light of the detailed description and with reference to the following figures.





BRIEF DESCRIPTION OF THE FIGURES


FIG. 1A is a schematic representation of the experimental design to analyze the proteome dynamics of tumors in a murine model of HeLa and SiHa cell lines.



FIG. 1B shows the kinetics of tumor growth of the HeLa (yellow) and SiHa (blue) cell lines. The kinetic endpoints (30, 45 and 50 days) were used to perform the proteomic analysis.



FIG. 1C shows that GSTM3 was identified in HeLa tumors and GSTP1 in SiHa tumors in 2-D electrophoresis. Expression levels of both proteins were confirmed by immunoblot analysis.



FIGS. 1D-1F show a representative image of each protein on days 30, 45 and 50. (1D) 14 proteins with constant expression in HeLa and SiHa tumors; (1E) 3 proteins with a subexpression over time in HeLa and SiHa; (1F) 17 proteins with different expression between HeLa and SiHa tumors.



FIGS. 2A-2C show an analysis made through the GeneCodis website of the enrichment of the gene ontology of the proteins identified in the tumor of HeLa and SiHa cells. (2A) Biological processes enriched in overregulated shared proteins; (2B) biological processes enriched in constant shared proteins; (2C) biological processes enriched in shared downregulated proteins.



FIGS. 3A-3F show that GSTM3 interacts with TRAF6 in cervical cancer tumors HeLa and SiHa, under physiological conditions. (3A) Cytoscape interaction network representing the prey interactions of GSTM3; (3B) co-immunoprecipitation analysis of the GSTM3 and TRAF6; (3C) Western spotting for TRFA6, ERK, pERK, NF-κ, PNF-κB; IKBα, p38, pp38, JNK, pJNK and TLR4 in the protein extracts of HeLa and SiHa tumors; (3D) Proportional Venn diagram of proteins secreted in cervical cancer cell lines with 264 common proteins; (3E) identification of two proteins secreted in vitro that can activate the TLR4 signal pathway; HSP60 and HSP70; (3F) Western spotting of TRL4 activators; HSP70 and HSP60 in cervical cancer tumors, HSP60 secreted in SiHa and HeLa tumors on day 50, and HSP70 protein secreted in SiHa tumors at 45 days and HeLa tumors at 30 and 50 days.



FIGS. 3G-3H show workflows to obtain secreted proteins in vivo or ex vivo.



FIGS. 4A-4F show that GSTM3 interacts with HPV18 E7 wherein GST and E7 provide survival advantages to cells exposed to stress conditions. (4A) The overlap of GSTP1 and GSTM3 proteins show high structural similarities (green-orange), unconserved structures (gray) and the structure of the HPV18 E7 protein (blue) using the HPV16 E7 as a mold; (4B) interaction of the human recombinant ScGSTM3 N-6× his-tag protein with the E7 of the HPV18 protein; (4C) HeLa cells were transfected with a plasmid expressing HE718C-6× his-tag; (4D) shows PAEP assays; (4E) survival test with 6.0 mM cisplatin; (4F) PAEP assays using the MDA cell line with 6.0 mM cisplatin.



FIGS. 4G-4H show that MDA-MB-231 is a negative cell line for HPV18 and GSTs proteins (GSTM3 and GSTP1) (4G), (4H) show the expression of GSTM3 and GSTP1 proteins in breast tumor cuts (of MDA) and colon cancer (COLO 237) generated in mice of the Nu/Nu strain.



FIGS. 4I-4L show the construction of yeast plasmids, transformation and expression of recombinant protein. (4I) Human recombinant protein of GSTM3 with a histidine (His) tag that was expressed in the yeast Saccharomyces cerevisiae; (4J) after capturing the recombinant GSTM3, it was incubated with a HeLa cell protein extract (positive for HPV18); (4K) HPV18 E7 recombinant protein with a His expression in the HeLa cell line; (4L) after capturing the E7 of the 6× his-tag recombinant HPV18 with nickel beads, it is incubated.



FIGS. 5A-5E show that “gene inhibition” (knock-down) of GSTM3 and GSTP1 affects the viability of cervical cancer cell lines in culture. (5A) Genetic experiments of “gene inhibition” (knock-down) of GSTM3 and GSTP1 in HeLa cells (yellow lines) and HaCaT (blue lines); (5B) viability assay with OAS-control or OAS-GST (at 640 ng/mL) determined by staining with violet crystal; (5C) live/dead cell assays determined by staining SYTO 9 (live cells, green color and dead, red cells) in cells treated with OAS-control and OAS-GST (640 ng/mL); (5D-E) GST inhibition with antisense oligonucleotide treatment in cervical cancer cell lines.



FIGS. 6A-6E show how “gene inhibition” (knock-down) of GSTM3 and GSTP1 affects tumor progression (TP) in cervical cancer tumors.



FIG. 6F shows the growth kinetics of cell lines with and without SFB indicating that there are no significant differences when the cells reach 70% confluence on the sixth day.



FIGS. 7A-7B illustrate that HeLa tumors treated with OAS-GSTM3 show inactivation of ERK and p65 NF-κB proteins.



FIGS. 7C-7D show that in CaLo tumors only pERK was inactivated after treatment with any of the antisense oligonucleotides for GST.



FIGS. 7E-7F show that for SiHa tumors only NF-κB was inactivated by any of the treatments.



FIGS. 7G-7H show that in CaSki tumors both proteins were inactivated after any treatment.



FIGS. 8A-8D shows the correlation between GST protein expression and survival of patients with cervical cancer. (8A) Representative specimens of invasive cervical cancer with different levels of expression of GSTM3 and GSTP1 (weak, moderate and high); (8B) ROI of GSTM3 and GSTP1 in 13 patients with cervical cancer evaluated; (8C) the percentage of ROI of GSTP1 was classified as weak (10%), moderate (20-50%) and high (51-100%); (8D) Kaplan-Meier survival graph, for the advanced stage of cervical cancer according to the levels of protein expression of GSTM3 and GSTP1 (log-rank test, p<0.05).



FIGS. 8E-8F shows the expression of GSTM3 and GSTP1 in cervical cancer in the terminal stage.



FIG. 9 shows that during the progression of cervical cancer several processes such as cell survival, proliferation and evasion of apoptosis through the MAPK kinase and NF-κB pathways are stimulated by the presence of GSTM3 and/or GSTP1. The “gene inhibition” (knock-down) of GSTM3 and GSTP1 affects the activation of activated apoptosis through the activation of JNK and p38 or the phosphorylated inhibition of NF-κB and ERK.





DETAILED DESCRIPTION OF THE INVENTION

As indicated above, the word cancer is a generic term that designates a broad group of diseases that can affect any part of the body, such as lung cancer, breast cancer, colorectal cancer, prostate cancer, stomach cancer, liver cancer, esophageal cancer, cervical cancer, thyroid cancer, bladder cancer, non-Hodgkin lymphoma, pancreatic cancer, leukemia, kidney cancer, uterine body cancer, oropharyngeal cancer, brain and central nervous system cancer, ovarian cancer, melanoma cancer, gallbladder cancer, larynx cancer, multiple myeloma cancer, nasopharyngeal cancer, laryngopharyngeal cancer, Hodgkin lymphoma, testicular cancer, salivary gland cancer, vulvar cancer, Kaposi sarcoma, penile cancer, mesothelioma, and vaginal cancer, among others, so it should be understood that a person skilled in the art will appreciate that the invention described below is susceptible to variations and modifications different from those specifically described and therefore, the present invention includes all such variations and modifications as well as all the stages, features, compositions and compounds referred to or indicated therein, whether in individual or collective and any of all combinations or any of two or more of the stages or features.


Furthermore, it should also be understood that the present invention is not limited in scope to the specific embodiments described therein, which are proposed for exemplification purposes only. Functionally equivalent products, compositions, combinations and methods as well as their application in mammals are clearly within the scope of the invention, as described.


Tumor progression (hereinafter also referred to as PT) involves changes in the deregulation of metabolic and cellular processes. The study of tumor proteome dynamics represents the protein changes of this deregulation mechanism during PT. Therefore, the study of proteome dynamics for example in cervical cancer (hereinafter also referred to as CC) will provide relevant information to understand PT and the disease to be treated.


Protein overexpression of the glutathione S transferase GSTM3 and GSTP1 genotypes during tumor progression (PT) plays a regulatory role through interaction with proteins, for example TRAF2/6 and therefore an evasion of transduction of signals of apoptosis activation, favoring cell survival and PT. In addition, GST expression is involved in the modulation of detoxification processes in cancer cells and, therefore, participates in the survival response to conventional chemotherapy in patients with CC and other types of cancer.


The present invention relates to antisense oligonucleotides of glutathione S transferase (GSTs) such as GSTM3 and GSTP1, as novel candidates for use as therapeutic targets and/or prognostic factors for patients (mammals) who have been diagnosed with CC and other types of cancer.


The identification of candidate subjects for the treatment of cancers and their treatment is carried out by identifying the protein expression of glutathione S transferase proteins (GSTs). Treatment includes the use of antisense oligonucleotides targeting the messenger ribonucleic acids (mRNA) of the GSTM3 and GSTP1, which give tumor cells greater resistance to chemotherapies.


The antisense oligonucleotide (OAS) is preferably any antisense oligonucleotide, which reduces the expression levels of the GSTMs and thus increases the sensitivity of the cell, tissue and/or organ to the chemotherapeutic agent in vitro, ex vivo, or in vivo.


In a first embodiment, the antisense oligonucleotide is an oligonucleotide composed of subunits called “nucleotides”, wherein each nucleotide is made up of three parts: a sugar or a functional analogue thereof, a nitrogen base and a functional group that serves as an internucleotide bond (usually a phosphate group) between the subunits that make up the oligonucleotide. Each of these components may contain the following modifications:




embedded image


wherein


X means a sugar, ribose in the case of RNA or deoxyribose in the case of DNA or the functional analog thereof in the nucleotide.


B means the nitrogenous base bound to sugar or a functional analogue thereof, and R means the functional group in carbon 2′ of sugar when it is ribose.


In a second embodiment, modifications to ribose sugar, without being limiting, are:


Modifications in the 2′ position of ribose sugar that include the replacement of the OH group by different groups among which are (2′-O-methyl (2′OMe), 2′-O-methoxyethyl (2′-OMOE), 2′fluor (2′-F) and 2′-chiral fluor (2′-F))




embedded image


wherein base refers to the nitrogenous base of the nucleotide.


In a third embodiment, modifications in ribose include the use of an analogue of bicyclic ribose, wherein the ribose structure contains an additional ring, for example, ribose with 2′-O, 4′-C-methylene (LNA-blocked nucleic acid), 2′-O, 4′-C-oxymethylene, 2′-O, 4′-C-methylene-β-D-ribofuranosil rings, among others.


Additionally, the substitution of ribose sugar can be carried out by analogous functional groups with similar function, for example, the substitution of the ribose by another sugar such as arabinose, morpholino, or by ribose analogs with a spirocyclic ring in different positions of the sugar ring, without being such limiting examples.


The following are examples of some of the different modifications to the sugary part of the oligonucleotide.




text missing or illegible when filed


In a fourth embodiment, modifications to the nucleobases or nitrogenous bases include, but are not limited to, adenine, cytosine, guanine, thymine, as well as their modifications 5-methylcytosine, 2-aminopurine, 2-amino-6-chloropurine, 2-diaminopurine, hypoxatin, 5-propynyl uracil, 2-thio thymine, N3-thioethyl thymine, 3-deaza adenine, 8-azido adenine and 7-deaza guanine, or the use of universal bases such as 3-nitropyrrole, imidazole-4-carboxamide, 5-nitroindole.


The following are examples of some structures of different types of modified nucleobases and nucleobase analogs.




embedded image


In a fifth embodiment, the modifications in the column or skeleton of the internucleoside groups, which bind them, can be but are not limited to:




embedded image


5′-N-carbamate, methylene-methylimine, amide, phosphorodithioate, thioether, thioformacetal and mercaptoacetamide. The substitution of the phosphodiester or phosphorothioate group can be carried out by phosphorodiamidate groups with piperazine and morpholino residues (PMOs).


For a person skilled in the art of the present invention it will be obvious that, within the scope, the chimeras resulting from the mixture of 2 or more chemical modifications mentioned above are also included, for example without being limiting, the replacement of the ribose by a morpholino ring and replacement of the phosphodiester group with phosphorodiamidate groups without charge (PMOs), the replacement of ribose and the phosphodiester bond is replaced by pseudopeptide N-(2 aminoethyl) glycine and the nucleobase is linked to the oligonucleotide column through of an ethyl enecarbonyl bond (PNA), a chimera where the ribose contains an alkyl substituent at the 2′ position and the phosphodiester bond is replaced by a phosphotiester bond, a chimera wherein the ribose is replaced by a ribose analogue with 2′-O,4′-C-methylene (LNA) rings and the phosphodiester bond is substituted by a phosphotiester bond, among others.


Examples of chimeras resulting from the combination of the different components of a nucleotide are by way of example:




embedded image


embedded image


DESCRIPTION OF THE PREFERRED EMBODIMENT
Tumor Progression Model

In order to create a suitable model to study tumor progression (PT), cervical cancer cell lines (SiHa and HeLa), triple negative breast (MDA-231-MB) and colon (COLO 205) were used to generate grafted tumors in mice. Cancer cells were cultured at 70% confluence and 107 cells were inoculated into female mice of the Nu/Nu strain for 4 to 6 weeks. The tumor volume was measured according to the equation of the volume of an ellipsoid described as: Vtm=η/6 (L*W*H), where L: length, W: width and H: height, and were taken in seven different moments of PT, for example, a kinetics of HeLa and SiHa tumors was taken at 5, 10, 15, 20, 30, 45 and 50 days after inoculation. The first four measurement times showed a low progression of tumor growth. However, at the endpoints of kinetics, tumor volume grew exponentially for HeLa cell tumors. From day 30 to 45 the average tumor volume doubled and by day 50 the average volume was 3 times more than the previous measurement. For SiHa cells, tumor growth rates were lower than HeLa; from 30 to 45 days, SiHa tumors grew 1.6 times more, while, in the last five days, tumors grew on average 1.6 times larger (see FIGS. 1A-B). Due to these results, the dynamics of PT were evaluated more at the level of proteome between times 30, 45 and 50 days after inoculation.


Analysis of 2D-PAGE Gels and Protein Identification.


Tumors of the two types of cancer cells (HeLa and SiHa) were collected and total proteins were extracted for analysis by means of two-dimensional electrophoresis gels (2D-PAGE). The analysis of the 2D-PAGE images of each repetition for each time studied and for each type of cell was carried out using the PDQuest software. An average of 866 electrophoretic entities (spots or “stains”) were detected for samples of HeLa tumors at each repetition. For SiHa tumors, the average of spots or “stains” detected in 2D-PAGE images was 766. The correlation coefficient between repetitions for each tumor time and cell type was determined from the 2D-PAGE maps.


Table 1 below shows that the correlation coefficient in all tumors was greater than 0.7 in both types of cells. The proteomic profile was obtained each time and then compared to find differentiated proteins during PT.











TABLE 1






HeLa
SiHa


Time (days)
Stains/CCf*
Stains/CCf*

















30
824/0.710
765/0.731


45
763/0.723
768/0.836


50
1012/0.724 
766/0.756





*CCf = correlation coefficient.






In addition, 601 points were detected in HeLa tumors in the three times of PT, while in SiHa tumors the number of common points was 716 (see FIG. 1A). For protein identification, a total of 90 tumor gel points was selected including both HeLa and SiHa cell types, based on their abundance patterns between the ages of the tumors. All electrophoretic or 2D gel entities were processed as described in the experimental section and identified after MALDI-TOF mass spectrometry analysis.


Table 2 below shows that 46 different proteins were identified from HeLa tumors, including 34 with constant expression through PT, 7 proteins showed a negative regulation along PT, 3 proteins increased their abundance during tumor growth, and 2 were found with an oscillating pattern.









TABLE 2







Total proteins expressed in HeLa tumors (days 30, 45 and 50)











HeLa
ID Swissprot *
MASCOT Score**















Constant
ENOA_HUMAN
92




ANXA1_HUMAN
134




ANXA3_HUMAN
148




ANXA5_HUMAN
65




ATPB_HUMAN
126




BGH3_HUMAN
135




CH60_HUMAN
74




CLIC1_HUMAN
80




CP21A_HUMAN
67




DHE3_HUMAN
150




EF2_HUMAN
178




EIF3I_HUMAN
81




FSCN1_HUMAN
126




GDIB_HUMAN
83




GRDN_HUMAN
73




GRP78_HUMAN
189




GSTO1_HUMAN
89




HSP71A_HUMAN
78




HSPB1_HUMAN
136




K1C17_HUMAN
102




K2C8_HUMAN
98




PDIA3_HUMAN
165




PGAM1_HUMAN
73




PHB_HUMAN
136




PKHA2_HUMAN
66




PRDX2_HUMAN
75




PRDX4_HUMAN
107




PSA1_HUMAN
73




PSA5_HUMAN
81




PSB4_HUMAN
85




RAN_HUMAN
73




RUVB1_HUMAN
85




TCPZ_HUMAN
109




TPIS_HUMAN
102



Downregulated
ACTS_HUMAN
172




ANXA2_HUMAN
68




DDX3X_HUMAN
104




IF4A1_HUMAN
134




TPM3_HUMAN
65




TPM4_HUMAN
106




VIME_HUMAN
171



Overexpressed
G3P_HUMAN
79




GSTM3_HUMAN
81




LDHB_HUMAN
106



Oscillation
HSP7C_HUMAN
134




TPM2_HUMAN
71







* Swiss-Prot database.



**MASCOT Score database






Table 3 below shows that a total of 44 proteins were identified from the SiHa cells. The identified proteins were distributed according to their expression pattern, 20 were found without differences in the three tumor ages evaluated, 8 decreased their abundance during PT, while 16 showed an increasing pattern. When analyzing all the proteins identified in tumors of both types of cells (Hela and SiHa), it was found that 34 proteins were shared between the two types of tumors, including 14 that show the same pattern of expression.









TABLE 3







Total proteins expressed in SiHa tumors (days 30, 45 and 50)











SiHa
ID Swissprot
MASCOT Score















Constant
ACTS_HUMAN
122




ACTG_HUMAN
107




ANXA1_HUMAN
132




ANXA5_HUMAN
102




BGH3_HUMAN
83




CH60_HUMAN
122




DHE3_HUMAN
80




EF2_HUMAN
184




FSCN1_HUMAN
108




HNRH3_HUMAN
153




HSP71_HUMAN
183




HSP7C_HUMAN
134




K1C17_HUMAN
192




LDHB_HUMAN
111




PCNA_HUMAN
114




PDIA3_HUMAN
135




PHB_HUMAN
131




PRDX4_HUMAN
80




PSB4_HUMAN
75




PSA1_HUMAN
90



downregulated
ATPB_HUMAN
174




DDX3X_HUMAN
134




GRP75_HUMAN
75




K1C17_HUMAN
176




RUVB1_HUMAN
137




TPM4_HUMAN
106




TCPZ_HUMAN
158




VIME_HUMAN
147



Overexpressed
ACTB_HUMAN
101




ANXA3_HUMAN
168




CLIC1_HUMAN
83




EIF3I_HUMAN
99




ENOA_HUMAN
81




ENOA_HUMAN*
114




GSTP1_HUMAN
74




GRP78_HUMAN
154




HSPB1_HUMAN
108




K1C17_HUMAN
154




K2C8_HUMAN
172




PGAM1_HUMAN
135




PRDX2_HUMAN
121




PSA5_HUMAN
96




RAN_HUMAN
113




TPIS_HUMAN
81







*Swiss-Prot database.



**MASCOT Score database






Among the proteins with levels of overexpression, two family members were identified Glutathione S-transferase (GSTM3 and GSTP1). GSTM3 was identified in HeLa tumors and GSTP1 in SiHa tumors (see FIG. 1C). Expression levels of both proteins were confirmed by immunoblot analysis (see FIG. 1C). In addition, immunoblotting revealed that both proteins with over-expression patterns are observed in SiHa tumors. However, in HeLa tumors only overexpression was confirmed for GSTM3, and it was found that GSTP1 is not detectable at any stage of the tumor.


Bioinformatic Analysis


Subsequently, the proteins identified in both tumors were used to perform a functional enrichment analysis based on the biological processes of gene ontologies (Gene Ontology: GO.


The proteins were grouped according to their expression levels and subjected to an enrichment analysis. The results indicated that proteins that increase their levels during PT are mainly involved in anti-apoptotic, cell division, glycolysis, angiogenesis, viral reproduction and regulation of apoptotic processes (see Checa-Rojas A., et al. GSTM3 and GSTP1: novel players driving tumor progression in cervical cancer, Oncotarget. 2018; 9: 21696-21714). In addition, including all identified proteins, the results suggest that during PT the overrepresented pathways are related to the cellular response to stress, the MAPK6/MAPK4 and NIK/NF-kappaB signaling pathways.


On the other hand, data mining analysis revealed that GSTM3 and GSTP1 interact with the proteins of factors associated with the tumor necrosis factor receptor (TRAF). However, said analysis can be extended to other proteins. Specifically, the interaction of GSTP1 with TRAF2 previously validated in HeLa cells (Wu, Y., Fan, Y., Xue, B., Luo, L., Shen, J., Zhang, S., . . . Yin, 2. (2006). Human glutathione S-transferase P1-1 interacts with TRAF2 and regulates TRAF2—ASK1 signals. Oncogene, 25, 5787-5800), and GSTM3 was reported as an interactor of TRAF6 (tumor necrosis factor receptor associated with factor 6) (Rouillard, A D, Gundersen, G W, Fernandez, N F, Wang, Z., Monteiro, C D, McDermott, M G, & Ma'ayan, A. (2016). The harmonizome: a collection of processed datasets gathered to serve and mine knowledge about genes and proteins. Database, 2016, baw100.) (see FIG. 3A).


To demonstrate that this interaction occurs under physiological conditions, TRAF6 expression was observed in both CC tumors (see FIG. 3B). It was observed that TRAF6 was expressed only in HeLa tumors. After that, the interaction analysis was carried out, by coimmunoprecipitation (IP). It was found that, GSTM3 co-immunoprecipitates with TRAF6 and vice versa (see FIG. 3B). It is shown that GSTM3 is associated with TRAF6 CC tumors.


Modulation of MAPK Signaling During PT


Given the importance of TRAF proteins on the downstream activation of the mitogen-activated protein kinase (MAPK) cascade, the phosphorylated version of NF-κB p65 (ser529), ERK, JNK and p38 by Western spotted (blot) of the PT proteins. The results showed that the phosphorylation of p38 and JNK was reduced over time in both CC tumors, but not in pNF-κB and pERK (see FIG. 3C). The results indicate that during PT the apoptotic processes are repressed and, therefore, cell proliferation is constantly activated via ERK and NF-κB. FIG. 3A shows the interaction network representing GSTM3 dam interactions, which can be visualized by the network limits. This analysis was performed to obtain the interactions reported by the SysBiomics database, in which it was observed that TRAF6 interacts with GSTM3. FIG. 3B shows the co-immunoprecipitation of GSTM3 and TRAF6. In the proportional Venn diagram of FIG. 3D the secreted proteins of the CC cell lines with 264 common proteins are were observed and FIG. 3E shows that two proteins identified in secreted proteins that can activate the TLR4 signal pathway expressed in vitro in HSP60 and HPS70. The western spotting of HSP70 and HSP60 activators of TRL4 in proteins secreted by CC tumors is shown in FIG. 3F. In addition, it is shown that HSP60 was expressed in SiHa and HeLa tumors at 50 days and HSP70 protein expressed in SiHa tumors at 43 days, and Hela tumors at 30 and 50 days.


Endogenous Secreted TLR4 Receptor Activators in the CC


On the other hand, it is known that the activation of the TLR4 pathway is driven by the presence of lipopolysaccharides (LPS) from bacterial infections, but also by endogenous activators such as HSP60 and HSP70 thermal shock proteins. To demonstrate that CC cell lines can express endogenous activators of the Toll type receptor 4 (TLR4), an in vitro analysis of the secreted proteins was performed using the HeLa and SiHa cell lines (see FIG. 3G). The following Tables 45 show the results obtained.


Secreted proteins were analyzed by LC-MS/MS and a total of 432 HeLa and 447 SiHa proteins were identified, of which 264 were common between both cell lines (see FIG. 3D). Among the endogenous activators reported for TLR4, two secreted proteins were identified as members of the family of thermal shock proteins, HSP60 and HSP70 for both cell lines (see FIG. 3E). To determine whether these proteins were also expressed during PT, the proteins ex vivo secreted in CC tumors were then analyzed by western spotting (see FIG. 3F, FIG. 3H). The results were correlated in in vivo and ex vivo experiments, indicating that secretion of HSP60 and HSP70 activates TLR4 signaling. See FIG. 3C.


Proteomic mass spectrometry data has been deposited in ProteomeXchange Consortium via the deposit of its associated PRoteomics IDEntifications (PRIDE) with the data set identifier PXD005466.









TABLE 4







Proteins secreted in HeLa cells















Matching


ID
Name
Mass
Score
peptides














P62258
14-3-3 epsilon protein
29155
321
8


P61981
14-3-3 gamma protein
28285
149
3


P63104
14-3-3 zeta/delta protein
27728
337
3


Q9NQ66
1-phosphatidylinositol-4,5-
0
51
4



bisphosphate



phosphodiesterase beta-1


Q4KWH8
1-phosphatidylinositol-4,5-
0
40
4



bisphosphate



phosphodiesterase eta-1


O75038
1-phosphatidylinositol-4,5-
0
37
4



bisphosphate



phosphodiesterase eta-2


P62081
40S ribosomal protein S7
22113
125
3


P08865
40S Ribosomal Protein SA
32833
38
2


P32754
4-hydroxyphenylpyruvate
44906
181
6



dioxygenase


P10809
60 KDa thermal shock protein,
61016
409
10



mitochondrial


P05388
60S P0 ribosomal acidic
34252
118
3



protein


P62899
60S ribosomal protein L31
14454
54
2


Q02878
60S ribosomal protein L6
32708
59
2


P52209
6-phosphogluconate
53106
245
2



dehydrogenase,



decarboxylation


P11021
Glucose-regulated 78 kDa
72288
751
1



protein


Q8IZT6
Microcephaly-associated
409540
52
6



protein similar to an



abnormal spindle


Q9BWD1
Acetyl-CoA
41324
77
1



acetyltransferase, cytosolic


P68032
Actin, alpha 1 heart muscle
41992
480
2


P68133
Actin, alpha skeletal muscle
42024
480
2


P62736
Actin, aortic smooth muscle
41982
93
2


P60709
Actin, cytoplasmic 1
41710
640
1


P63261
Actin, cytoplasmic 2
41766
640
1


P25054
Adenomatous polyposis coli
311455
47
6



protein


O00468
Agrina
214706
43
7


Q2M3C7
SPHKAP A-kinase anchor
186339
33
4



protein


P12814
alpha-actinin-1
102993
579
13


P35609
alpha-actinin-2
103788
34
2


O43707
alpha-actinin-4
104788
1436
6


P06733
alpha-enolase
47139
1172
6


Q96L96
alpha-protein kinase 3
201148
39
5


Q8TCU4
Alstrom 1 syndrome protein
0
47
7


Q01484
Anquirin-2
429990
35
5


Q12955
Anquirin-3
480113
36
6


P04083
Annex A1
38690
49
1


P07355
Annex A2
38580
471
10


P08758
Annex A5
35914
251
5


P46013
KI-67 antigen
358474
57
5


O00203
AP-3 beta-1 subunit complex
0
34
3


P04114
Apolipoprotein B-100
515283
64
5


O43150
Arf-GAP with SH3 domain, ANK
0
36
2



repeat and protein 2



containing the PH domain


P00966
Argininosuccinate synthase
46501
277
9


P17174
Aspartate aminotransferase,
46219
388
8



cytoplasmic


Q7Z591
Transcription factor
155044
36
3



containing AT connection


Q5T9A4
3B protein containing AAA
0
46
4



domain of the ATPase family


Q96QE3
Protein 5 containing the AAA
0
38
4



domain of the ATPase family


Q9BZC7
Member 2 of subfamily A of
269701
36
6



the cassette linking ATP


Q8IZY2
Member 7 of subfamily A of
234201
37
3



the cassette linking ATP


P53396
ATP-citrate synthase
120762
585
7


98160
Heparan basement membrane
468532
43
4



proteoglycan sulfate core



protein


Q562R1
Protein 2 similar to beta-
0
256
1



actin


P25098
1 beta-adrenergic kinase
0
34
3



receptor


P13929
Beta-enolase
46902
216
2


Q96T60
polynucleotide
0
32
4



phosphatase/bifunctional



kinase


Q8NFC6
Biorientation of chromosomes
0
42
6



in protein 1 of cell-similar



division


O14514
Brain specific angiogenesis
0
40
4



inhibitor 1


O60241
Brain specific angiogenesis
0
33
3



inhibitor 2


O60242
Guanine nucleotide exchange
201909
32
4



protein 2 inhibited Brefeldin



A


O60243
Brevican core protein
99056
43
2


O60244
Protein 1 containing WD and
0
64
4



Bromodomain repetition


O60245
Carbamoyl phosphate synthase
164835
133
3



[ammonia], mitochondrial


O60246
Cathepsin D
44524
220
6


O60247
Cathepsin Z
33846
167
4


O60248
ABL1 and CDK5 enzyme substrate
0
41
1


O60249
Protein 2 associated with CDK5
0
35
3



regulation subunit


O60250
Centlein
161504
47
3


O60251
F centromer protein
367537
54
6


O60252
Protein E associated with
0
54
5



centromer


O60253
Protein 350 associated with
350716
45
5



centrosome


O60254
Chlorine intracellular channel
26906
128
2



protein 1


O60255
O-Acetyltransferase coline
82483
33
4


O60256
CLIP association protein 1
0
66
4


O60257
Clusterin
52461
205
3


O60258
Cofilin-1
18491
195
3


O60259
Protein 17 containing double
0
37
2



helix domain


O60260
Protein 78 containing double
0
42
3



helix domain


O60261
Protein 87 that contains
0
44
4



double helix domain


O60262
Protein 93 containing double
0
39
4



helix domain


O60263
Alpha-1 (III) collagen chain
138479
33
4


O60264
Alpha-1 (VII) collagen chain
295041
57
7


O60265
Alpha-1 (XII) collagen chain
332941
1361
31


O60266
Alpha-1 (XIV) collagen chain
0
47
1


O60267
Collagen alpha-1 (XXVII) chain
0
45
7


O60268
Alpha-2 (XI) collagen chain
171670
61
10


O60269
Alpha-3 (VI) collagen chain
0
37
2


O60270
Cortactin Binding Protein 2
0
60
3


O60271
Protein 1 containing domain
388621
40
3



CUB and Sushi


O60272
Cubilin
398480
47
5


O60273
Dissociated protein 1 from
136289
174
1



NEDD8 associated with Cullin


O60274
Cystatin-C
15789
54
1


O60275
Heavy chain 1 of dynein 1
532072
54
6



cytoplasmic


O60276
Protein Cytokinesis Dedicator
0
50
3



6


O60277
Dermcidin
11277
66
1


O60278
Desmoplakin
331569
49
4


O60279
Dihydrolipoyl dehydrogenase,
54143
177
6



mitochondrial


O60280
Homologous B of protein 2
0
32
3



that interacts with Disco


O60281
DNA polymerase theta
197474
35
4


O60282
Catalytic subunit of DNA
0
43
5



polymerase zeta


O60283
DNA-dependent protein kinase
468788
65
9



catalytic subunit


O60284
Member 13 of DnaJ homolog
0
49
4



subfamily


O60285
Coupling protein 6
0
36
2


O60286
Heavy chain 10 of dieine,
514512
46
6



axonemal


O60287
11 dyneine heavy chain,
520711
55
6



axonemal


O60288
2 dyneine heavy chain,
0
35
4



axonemal


O60289
Heavy chain 3 of dynein,
470468
46
5



axonemal


060290
5 dyneine heavy chain,
528684
47
5



axonemal


O60291
6 dyneine heavy chain,
475679
35
7



axonemal


O60292
8 dyneine heavy chain,
514335
84
5



axonemal


O60293
Protein 1 containing dyneine
0
42
5



heavy chain domain


O60294
Distonin
0
50
4


O60295
Distroglycan
97381
74
1


O60296
Dystrophin
0
34
3


O60297
P400 protein that binds E1A
343276
35
7


O60298
E3 SUMO-protein ligase RanBP2
357974
68
5


O60299
E3 ubiquitin-protein ligase
526895
55
5



HERC2


O60300
E3 ubiquitin-protein ligase
0
46
4



HUWE1


O60301
E3 ubiquitin-protein ligase
0
52
2



UBR4


O60302
Protein similar to 6
0
37
4



associated with Echinoderm



microtubule


O60303
Elongation factor-alpha 1
50109
285
4


O60304
Gamma elongation factor 1
50087
185
1


O60305
Elongation phantom 2
95277
498
4


O60306
Eukaryotic initiation
46125
58
1



spectrum 4A-I


O60307
Eukaryotic initiation factor
46373
58
1



4A-II


O60308
ERF-3B subunit linking
0
32
1



eukaryotic peptide chain GTP



factor


O60309
Exophilin-5
0
40
6


O60310
FRAS1 extracellular matrix
442646
36
6



protein


O60311
Ezrin
69370
804
6


O60312
Anemia of Fanconi group I
149229
40
4



protein


O60313
Farnesyl pyrophosphate
48245
256
2



synthase


O60314
Fascin
54496
253
7


O60315
Fatty acid synthase
273254
853
23


O60316
Protein 1 containing type III
0
43
7



fibronectin domain


O60317
Protein 2 that interacts with
780119
44
6



fibrous sheath


O60318
Filagrine
434922
45
10


O60319
Filamine-B
277990
90
5


O60320
Fructose bisphosphate
39395
813
18



aldolase A


O60321
Fructose Bisphosphate
39431
190
4



Aldolase C


O60322
Galectin-1
14706
44
1


O60323
Protein that binds galectin-3
65289
628
17


O60324
Gamma-enolase
47239
221
2


O60325
Girdin
0
37
3


O60326
Glial fibrillary acidic
49850
81
2



protein


O60327
Glucokinase
0
36
3


O60328
Glucose-6-phosphate isomerase
63107
892
6


O60329
Glutamate receptor 3A subunit
125385
48
4



[NMDA]


O60330
Glutathione S-transferase
27548
76
2



omega-1


O60331
Glyceraldehyde-3-phosphate
36030
572
2



dehydrogenase


O60332
Alpha chain glycoprotein
13066
47
2



hormones


O60333
98 G-protein coupled receptor
0
37
4


O60334
GTP-binding nuclear Ran
24408
179
4



protein


O60335
Subunit of beta-2 protein
35055
32
2



similar to 1 nucleotide



binding of Guanine


O60336
70 kDa 1A/1B protein from
70009
639
2



thermal shock


O60337
Protein 1 of 70 kDa of
70331
345
1



similar thermal shock


O60338
Protein 6 of 70 kDa of
70984
253
2



thermal shock


O60339
71 kDa protein with thermal
70854
661
1



shock


O60340
Beta-1 thermal shock protein
22768
79
4


O60341
HSP 90-alpha thermal shock
84607
874
12



protein


O60342
HSP 90-beta thermal shock
83212
1010
10



protein


O60343
Hemicentin-1
613001
42
6


O60344
Hemicentin-2
542265
47
5


O60345
Hemoglobin alpha subunit
15248
56
2


O60346
Beta subunit of hemoglobin
15988
38
1


O60347
Hepatoma-derived growth
26772
63
2



factor


O60348
heterogenic nuclear
0
46
1



ribonucleoprotein A0


O60349
heterologous nuclear A1
38723
113
1



ribonucleoprotein


O60350
ribonucleoprotein A1-similar
34204
94
1



to 2 nuclear heterogeneous


O60351
heterogeneous nuclear
39571
57
1



ribonucleoprotein A3


O60352
heterogeneous nuclear D0
38410
114
3



ribonucleoprotein


O60353
heterogeneous nuclear A2/B1
37407
217
3



ribonucleoproteins


O60354
Histone H2A type 1
14083
134
1


O60355
Histone H2A type 1-B/E
14127
134
1


O60356
Histone H2A type 1-C
14097
134
1


O60357
Histone H2A type 1-D
14099
134
1


O60358
Histone H2A type 1-H
13898
134
1


O60359
Histone H2A type 1-J
13928
134
1


O60360
Histone H2A type 2-A
14087
134
1


O60361
Histone H2A type 2-B
13987
58
1


O60362
Histone H2A type 2-C
13980
134
1


O60363
Histone H2A type 3
14113
134
1


O60364
Histone H2A.J
14011
134
1


O60365
Histone H2A.x
15135
58
1


O60366
Histone H2B type 1-H
13884
100
4


O60367
Histone H2B type 2-F
13912
100
4


O60368
Histone H3.1
15394
158
6


O60369
Histone H3.1t
15499
155
6


O60370
Histone H3.2
15379
158
6


O60371
Histone H3.3
15318
160
6


O60372
Histone H3.3C
15204
44
1


O60373
Histone H4
11360
178
7


O60374
Histone-lysine n-
593017
49
6



methyltransferase MLL2


O60375
Histone-lysine N-
0
42
6



methyltransferase MLL3


O60376
Histone-lysine N-
0
39
4



methyltransferase SETD1A


O60377
Histone-lysine N-
0
42
5



methyltransferase, H3 lysine-



36 and H4 lysine-20 specific


O60378
Cutting protein-similar to 1
0
38
3



homeosequence


O60379
Protein homolog that induces
0
60
4



hydrocephalus


O60380
Protein that binds IgGFc
571639
40
3


O60381
Above that degrades insulin
0
36
3


O60382
Integrin alpha-10
0
32
4


O60383
Factor 3 linking interleukin
95279
48
3



enhancer


O60384
Intraflagelar transport
0
43
4



protein 81 counterpart


O60385
Keratin, type I cuticular Ha5
0
34
2


O60386
Keratin, type I cytoskeletal
58792
962
11



10


O60387
Keratin, type I cytoskeletal
53478
75
2



12


O60388
Keratin, type I cytoskeletal
49557
91
3



13


O60389
Keratin, type I cytoskeletal
51529
252
4



14


O60390
Keratin, type I cytoskeletal
49181
114
3



15


O60391
Keratin, type I cytoskeletal
51236
184
4



16


O60392
Keratin, type I cytoskeletal
48076
123
4



17


O60393
Keratin, type I cytoskeletal
48029
103
3



18


O60394
Keratin, type I cytoskeletal
44079
70
2



19


O60395
Keratin, type I cytoskeletal
49287
73
3



25


O60396
Keratin, type I cytoskeletal
49792
68
3



27


O60397
Keratin, type I cytoskeletal
50536
149
4



28


O60398
Keratin, type I cytoskeletal
62027
1295
12



9


O60399
Keratin, type II cuticular
64801
85
3



Hb4


O60400
Keratin, type II cytoskeletal
65999
1475
16



1


O60401
Keratin, type II cytoskeletal
61864
217
3



1b


O60402
Keratin, type II epidermal
65393
1547
13



cytoskeletal 2


O60403
Keratin, type II oral
65800
185
4



cytoskeletal 2


O60404
Keratin, type II cytoskeletal
64378
177
3



3


O60405
Keratin, type II cytoskeletal
57250
195
4



4


O60406
Keratin, type II cytoskeletal
62340
352
6



5


O60407
Keratin, type II cytoskeletal
60008
300
5



6A


O60408
Keratin, type II cytoskeletal
60030
330
4



6B


O60409
Keratin, type II cytoskeletal
51354
124
3



7


O60410
Keratin, type II cytoskeletal
57256
101
4



71


O60411
Keratin, type II cytoskeletal
55842
97
3



72


O60412
Keratin, type II cytoskeletal
58887
157
4



73


O60413
Keratin, type II cytoskeletal
57830
118
4



74


O60414
Keratin, type II cytoskeletal
59524
140
3



75


O60415
Keratin, type II cytoskeletal
0
42
3



78


O60416
Keratin, type II cytoskeletal
57800
181
4



79


O60417
Keratin, type II cytoskeletal
53671
131
4



8


O60418
Keratin, type II cytoskeletal
50494
78
2



80


O60419
Close relationship of IRRE-
0
46
3



similar to protein 2


O60420
Kinesin-like protein KIF17
0
34
2


O60421
Kinesin-like protein KIF18B
0
35
4


O60422
Kinesin-like protein KIF20B
0
58
2


O60423
Kinesin-like protein KIF26A
194468
47
2


O60424
Laminin alpha-1 subunit
336867
34
5


O60425
Laminin alpha-5 subunit
0
32
5


O60426
Laminin gamma-1 subunit
177489
35
2


O60427
Lethal malignant brain tumor
0
38
4



(3)-similar to protein 3


O60428
Protein kinase 2
0
48
3



serine/threonine rich in



leucine repetition


O60429
16A protein containing
151462
44
5



leucine-rich repeats


O60430
Receptive lipopolysaccharide
0
34
5



and anchor protein similar to



beige


O60431
L-lactate dehydrogenase A
36665
534
4



chain


O60432
L-lactate dehydrogenase A-
36484
55
2



similar to 6A


O60433
L-lactate dehydrogenase A-
41916
120
4



similar to 6B


O60434
L-lactate dehydrogenase B
36615
552
3



chain


O60435
1B protein related to low
515159
45
7



density lipoprotein receptor


O60436
Protein 4 related to low
0
38
5



density lipoprotein receptor


O60437
Protein 6 related to low
180314
39
3



density lipoprotein receptor


O60438
Lysine specific demethylase
175545
37
3



5B


O60439
Lysine specific histone
92039
37
5



demethylase 1B


O60440
Malate dehydrogenase,
36403
351
5



cytoplasmic


O60441
Malate dehydrogenase,
35481
113
3



mitochondrial


O60442
Protein associated with gene
0
47
6



MAX


O60443
Metalloproteinase Inhibitor 1
23156
90
2


O60444
Methylcytosine dioxygenase
0
49
4



TET1


O60445
Factor 1 reticulating
669721
50
7



microtubule, isoform 4


O60446
Microtubule-actin
0
44
5



crosslinking factor 1,



1/2/3/5 isoforms


O60447
Microtubule-associated
143049
49
5



serine/threonine protein



kinase 3


O60448
Suppressor tumor candidate 2
0
53
5



associated with microtubule


O60449
Midasin
0
62
10


O60450
Moesin
67778
719
6


O60451
Mucin-16
0
59
5


O60452
Mucin-19
597790
37
4


O60453
Multiple epidermal growth
161072
38
5



factor-similar to protein 6



domains


O60454
Protein 2 associated with
29394
36
3



multiple myeloma tumor


O60455
Miosin-14
227863
38
2


O60456
Miosin-3
223766
39
4


O60457
Miosin-7B
221251
41
6


O60458
Miosin-XV
395044
41
6


O60459
Nck associated protein 5
208409
41
6


O60460
Nebulin
0
44
5


O60461
Nesprin-1
1010398
39
8


O60462
Neurobeachin-similar to
0
39
4



protein 2


O60463
AHNAK protein associated with
628699
76
9



neuroblast differentiation


O60464
Neurofibromin
0
37
4


O60465
Notch protein 3 counterpart
0
37
4



of neurogenic site


O60466
Neuron browser 1
0
43
4


O60467
Pentraxin-1 neuronal
47093
240
10


O60468
Protein 1 of nuclear mitotic
238115
81
6



apparatus


O60469
Nucleoside diphosphate kinase
17138
348
9



A


O60470
Nucleoside diphosphate kinase
17287
284
7



B


O60471
BPTF subunit factor
338054
36
5



remodeling nucleosome


O60472
Obg-similar to ATPase 1
44715
118
1


O60473
Obscurin
867940
45
6


O60474
Defective partition 6 gamma
0
33
3



homologue


O60475
Peptidyl-prolyl cis-trans
18001
285
3



isomerase A


O60476
Peptidyl-prolyl cis-trans
51772
278
7



isomerase FKBP4


O60477
Pericentrin
0
39
6


O60478
Peroxidasin Homolog
0
34
4


O60479
Peroxiredoxin-1
22096
317
2


O60480
Peroxiredoxin-2
21878
378
8


O60481
Peroxiredoxin-4
30521
205
5


O60482
Peroxiredoxin-6
25019
237
5


O60483
Protein 1 that binds
21044
252
4



phosphatidylethanolamine


O60484
Beta subunit containing
0
40
3



phosphatidylinositol-4-



phosphate 3-kinase C2 domain


O60485
Phosphoglucomutase-1
61411
240
6


O60486
Phosphoglycerate kinase 1
44586
668
1


O60487
Phosphoglycerate mutase 1
28786
135
5


O60488
Phosphoserine
40397
128
3



aminotransferase


O60489
Member 2 of family G containing
147877
39
4



Pleckstrin homology domain


O60490
Plectin
531466
93
5


O60491
Plexin-A1
210933
44
4


O60492
1-similar protein to 1 of
0
55
4



polycystic kidney disease


O60493
Polycystin-1
0
53
5


O60494
Polyubiquitin-B
25746
158
3


O60495
Polyubiquitin-C
76982
148
3


O60496
Family member E of domain
121286
444
4



POTE anchirine


O60497
Prelamin-A/C
74095
123
2


O60498
Likely ATP DDX41-dependent
0
35
3



RNA helicase


O60499
Likely ATP-dependent DDX60-
0
35
2



similar RNA helicase


O60500
Likely ubiquitin-similar E3
0
37
3



HERC1 ligase protein


O60501
Likely MYCBP2 protein ligase
509759
60
5



from E3 ubiquitin


O60502
Likely terminal hydrolase
0
33
4



FAF-Y of terminal ubiquitin



carboxyl


O60503
Profilin-1
15045
179
4


O60504
Protein 1 related to density
504276
46
9



tested lipoprotein receptor


O60505
Proprotein convertase
74239
340
5



subtilisin/kexin type 9


O60506
Proteasome subunit alpha
29537
188
7



type-1


O60507
Proteasome subunit alpha
25882
178
6



type-2


O60508
Proteasome subunit alpha
28415
196
5



type-3


O60509
Proteasome subunit alpha
27382
303
8



type-6


O60510
Proteasome subunit alpha
27870
464
9



type-7


O60511
Proteasome beta subunit type-
26472
125
3



1


O60512
Proteasome subunit beta type-
22933
150
5



3


O60513
Proteasome beta subunit type-
29185
93
3



4


O60514
Proteasome subunit beta type-
28462
355
7



5


O60515
Proteasome subunit beta type-
25341
155
3



6


O60516
AHNAK2 protein
0
38
5


O60517
Arginine N-methyltransferase
0
59
1



3 protein


O60518
Bassoon protein
416214
46
3


O60519
Daple protein
228091
37
5


O60520
Disulfide isomerase protein
57081
98
5


O60521
Disulfide-isomerase A3
56747
635
13



protein


O60522
A6 disulfide isomerase protein
48091
172
3


O60523
FAM179A protein
111084
33
3


O60524
Protein counterpart “bassoon”
0
34
6


O60525
Irregular protein-2
133277
42
2


O60526
“Flutin” protein
566309
44
4


O60527
Shroom2 protein
176302
34
6


O60528
Shroom3 protein
216724
45
1


O60529
Companion protein-1
0
34
3


O60530
SZT2 protein
0
38
6


O60531
TANC1 protein
0
46
5


O60532
Protein-arginine deiminase
74618
42
1



type-1


O60533
Protocadherina Fat 1
0
33
6


O60534
Protocadherina Fat 3
505209
35
4


O60535
Purine nucleoside
32097
127
4



phosphorylase


O60536
Putative beta-actin-like
41989
230
5



protein 3


O60537
Putative elongation factor 1-
50153
285
4



alpha-similar to 3


O60538
HSP 90-beta 2 protein from
44321
237
3



putative thermal shock


O60539
Putative heterogeneous
34202
113
1



nuclear A1 ribonucleoprotein-



similar to 3


O60540
ASXL3 protein from the
241767
37
4



putative Polycomb group


O60541
Putative tubulin similar to
27534
39
2



alpha-4B protein


O60542
Isozymes M1/M2 pyruvate
57900
661
1



kinase isozymes M1/M2


O60543
Rab GDP alpha dissociation
50550
44
2



inhibitor


O60544
Rab GDP dissociation beta inhibitor
50631
549
3


O60545
Radixin
68521
524
3


O60546
Ras-related Rab-28 protein
0
37
1


O60547
Tyrosine-protein phosphatase
224161
35
3



beta receptor-type


O60548
Regulation of protein 1 of
188956
38
6



synaptic exocytosis


O60549
Retinitis pigmentosa 1-
0
43
3



similar to protein 1


O60550
Protein 23 that activates Rho
0
37
6



GTPase


O60551
Factor 2 homolog of Ribosome
35560
41
4



production


O60552
Ribosome binding protein 1
0
47
6


O60553
Rootletin
228388
49
7


O60554
Ryanodine receptor 2
564206
52
7


O60555
Sacsin
0
48
7


O60556
Splicing factor 1 rich in
27728
47
2



serine/arginine


O60557
Serine/threonine-protein
143903
34
3



kinase 36


O60558
Serine/threonine-protein
0
35
4



kinase ATR


O60559
Serine/threonine-protein
0
44
5



kinase SMG1


O60560
Serine/threonine-protein
0
50
4



kinase WNK2


O60561
Serpin B6
42594
160
4


O60562
Serpin H1
46411
145
5


O60563
Serum albumin
69321
267
4


O60564
SH3 and multiple protein
0
43
3



domain 2 repeat ankyrins


O60565
SH3 and multiple protein 3
0
44
6



domain repeat ankyrins


O60566
Signal transducer and
0
46
3



transcription activator 2


O60567
Spectrin alpha chain, brain
284364
343
4


O60568
Spectrin beta chain, brain 3
0
125
3


O60569
Spectrin beta chain, brain 4
0
39
5


O60570
Spliceosoma RNA helicase BAT1
48960
51
2


O60571
Stabilin-1
275300
36
5


O60572
Protein 1 containing - domain-
0
55
2



like - Sushi, nidogen and EGF.


O60573
Protein 1 containing Sushi,
0
39
5



von Willebrand factor type A,



EGF and pentraxin domain


O60574
Protein targeting Xklp2
0
33
2


O60575
Component 1 of telomerase
290307
36
4



protein


O60576
Tenascin-X
0
36
6


O60577
Tensin-1
185586
69
2


O60578
Thioredoxin
11730
187
1


O60579
Thioredoxin reductase 1,
70711
392
10



cytoplasmic


O60580
Titin
3813810
124
24


O60581
Toll-similar to receptor 10
0
40
2


O60582
C10orf93 protein containing
0
34
4



TPR repeat


O60583
Transcription factor TFIIIB
293705
35
7



component B “homologous


O60584
Transferrin receptor protein 1
84818
896
4


O60585
Protein associated with
437318
52
7



domain



transformation/transcription


O60586
Protein 2 containing acidic
309237
36
6



double helix transformation


O60587
ATPase transitional
89266
338
1



endoplasmic reticulum


O60588
Transketolase
67835
965
5


O60589
Treacle protein
0
42
5


O60590
Triosephosphate isomerase
26653
703
16


O60591
Triple functional domain
0
51
4



protein


O60592
Alpha-1B tubulin chain
50120
715
15


O60593
Alpha-1C tubulin chain
49863
715
15


O60594
Alpha-4A tubulin chain
49892
39
2


O60595
Alpha-8 tubulin chain
50062
39
2


O60596
Beta tubulin chain
49639
442
9


O60597
Beta-3 tubulin chain
50400
245
5


O60598
Beta-8 tubulin chain
49744
102
3


O60599
Ribosomal ubiquitin-40S S27a
17953
167
3



protein


O60600
L40 protein from ribosomal
14719
167
3



ubiquitin-60S


O60601
Uncharacterized C4orf37
50628
32
3



protein


O60602
KIAA0802 protein without
0
50
6



characterization


O60603
KIAA1109 protein without
0
59
6



characterization


O60604
KIAA1614 protein without
0
40
6



characterization


O60605
UPF0556 C19orf10 protein
18783
218
7


O60606
Urotensin-2
0
34
1


O60607
Utrophin
394220
38
4


O60608
13D protein associated with
491535
44
4



vacuolar protein


O60609
Vinculin
123722
358
9


O60610
Von Willebrand factor
309058
38
5


O60611
Protein 3 containing WD
0
38
5



repeat and FYVE domain


O60612
Protein 35 containing WD
0
36
2



repeating


O60613
KIAA1875 protein containing
180192
38
4



WD repeat


O60614
Protein 2 containing Xin
0
36
5



Actin binding repeater


O60615
Protein 13 containing CCCH
0
44
6



zinc finger domain


O60616
Zinc finger protein 142
187758
34
3


O60617
Zinc finger 469 protein
409949
44
5
















TABLE 5







Proteins secreted in SiHa cells.















Matching


ID
Name
Mass
Score
peptides














P31946
14-3-3 beta/alpha protein
28307
78
3


P62258
14-3-3 epsilon protein
29486
287
5


P27348
14-3-3 theta protein
28128
75
3


P63104
14-3-3 zeta/delta protein
28011
142
4


Q9NQ66
1-phosphatidylinositol-4,5-
0
44
3



bisphosphate



phosphodiesterase beta-1


Q4KWH8
1-phosphatictylinositol-4,5-
0
39
2



bisphosphate phosphodiesterase eta-1


P43686
26B Protease Regulatory 6B
47644
96
3



Subunit


P62269
S18 40S ribosomal protein
17740
73
3


P62847
S24 40S ribosomal protein
15509
61
1


P62081
S7 40S ribosomal protein
22145
119
4


P08865
SA 40S ribosomal protein
0
37
4


P32754
4-hydroxyphenylpyruvate
45256
313
7



dioxygenase


P10809
60 kDa thermal shock
61478
288
8



protein, mitochondrial


P05388
P0 60S acidic ribosomal
34567
229
5



protein


P62913
L11 60S ribosomal protein
20516
114
2


P30050
L12 60S ribosomal protein
18011
51
1


P83731
L24 60S ribosomal protein
17930
46
1


P52209
6-phosphogluconate
53852
257
4



dehydrogenase,



decarboxylation


P11021
78 kDa regulated glucose
72580
410
9



protein


Q8TE58
A disintegrine and
106076
37
1



metalloproteinase with



thrombospondin motifs 15


Q8IZT6
Associated protein-similar
414501
44
6



to abnormal microcephaly-



spindle


Q9BWD1
Acetyl-CoA
42030
70
2



acetyltransferase, cytosolic


P68032
Actin, heart muscle 1 alpha
42596
280
12


P68133
Actin, alpha skeletal muscle
42644
280
12


P62736
Actin, aortic smooth muscle
42644
108
2


P60709
Actin, cytoplasmic 1
42330
721
19


P63261
Actin, cytoplasmic 2
42386
721
19


P53999
Transcriptional p15 co-
14466
41
1



activator of activated RNA



polymerase


P23526
Adenosylhomocysteinase
48521
188
5


P00568
Adenylate kinase isoenzyme 1
21815
69
2


Q01518
Protein 1 associated with
52493
62
2



adenylyl cyclase


P84077
ADP ribosylation factor 1
20838
48
2


P61204
ADP-ribosylation factor 3
20742
48
2


P84085
ADP-ribosylation factor 5
20727
48
1


P12814
alpha-actinin-1
104005
340
12


O43707
alpha-actinin-4
105636
483
15


P06733
alpha-enolase
47631
885
10


Q8TCU4
Alstrom syndrome protein 1
463685
50
5


Q9BXX3
Protein 30A containing
161035
43
5



Ankyrin repeat domain


Q53LP3
Protein 57 containing repeat
56003
36
3



domain of Ankyrin


Q12955
Ankyrin-3
483809
43
6


P07355
Annex A2
38952
330
6


P08758
Annex A5
36099
145
3


P46013
KI-67 antigen
361481
62
5


P04114
Apolipoprotein B-100
517939
64
6


O43150
Arf-GAP with SH3 domain,
0
40
3



protein 2 containing PH



domain and ANK repeat


P00966
Argininosuccinate synthase
46935
310
10


P54136
Arginyl-tRNA synthetase, cytoplasmic
76415
33
3


P15848
Arilsulfatase B
0
36
2


P17174
Aspartate aminotransferase,
46543
111
4



cytoplasmic


Q7Z591
Transcription factor
156176
56
4



containing AT hitch


Q5T9A4
3B protein containing AAA
0
41
5



domain of the ATPase family


Q96QE3
Protein 5 containing AAA
209929
35
3



domain of the ATPase family


Q86UQ4
Member 13 of cassette
582507
46
4



subfamily A linking ATP


Q8IZY2
Member 7 of cassette
236929
40
3



subfamily A linking ATP


P53396
ATP-citrate synthase
122234
657
3


Q03989
5A protein containing
0
39
2



interactive domain rich in



AT


P98160
Base heparan sulfate
479940
37
3



proteoglycan basement



membrane specific protein


Q5H9F3
Co-repressor BCL-6-similar
0
33
4



to protein 1


Q13884
Beta-1-Sintrophin
0
35
3


Q562R1
Beta-actin-like protein 2
42596
156
4


P13929
Beta-enolase
47394
192
3


Q8NFC6
Biorientation of chromosomes
0
35
4



in protein 1 of cell-similar



division


O60241
Brain specific angiogenesis
0
52
7



inhibitor 2


Q9NYQ6
Receiver 1 type G seven
335176
71
5



steps of EGF LAG chain


Q9NYQ7
Receiver 3 type G seven
363521
32
4



steps of EGF LAG chain


Q9H251
Caderin-23
0
39
6


Q8N3K9
Protein 5 associated with
451703
43
4



cardiomyopathy


Q8WXD9
Caskin-1
0
33
4


Q9UBR2
Cathepsin Z
34610
136
3


Q14004
Protein kinase 13 cell
0
38
3



division


Q9HC77
Centromer J protein
0
34
4


Q02224
Centromere associated
0
36
3



protein E


Q5VT06
350 protein associated with
0
37
4



centrosome


O00299
Chloride intracellular
27280
136
3



channel protein 1


Q12873
Chromodomain-helicase-DNA
0
37
6



binding protein 3


Q00610
1 heavy clatrin chain
193946
143
10


Q7Z460
CLIP association protein 1
0
35
4


P10909
Clusterin
53287
115
2


Q9UBF2
Coatomer subunit gamma-2
0
32
4


P23528
Cofilin-1
18783
245
4


Q02388
Alpha-1 collagen chain (VII)
296298
46
4


Q99715
Alpha-1 collagen chain (XII)
334878
493
26


P39060
Alpha-1 collagen chain
0
42
5



(XVIII)


P08123
Alpha-2 collagen chain (I)
129917
34
4


P13942
Alpha-2 collagen chain (XI)
0
60
4


P12111
Alpha-3 collagen chain (VI)
345759
42
6


Q8WZ74
Cortosterin Binding Protein
183844
37
3


P12277
Creatine kinase type B
43083
39
2


Q86VP6
Dissociated protein 1 from
138525
51
2



NEDD8 associated with cullin


P01034
Cystatin-C
16081
128
2


Q14204
Heavy chain 1 of
536760
48
6



cytoplasmic dinneine 1


Q96HP0
Protein Cytokinesis
0
40
4



Dedicator 6


O43598
Deoxyribonucleoside 5′-N-
19259
65
1



glycosidase monophosphate


O14531
Protein 4 related to
0
41
4



dihydropyrimidinase


Q14689
Protein 2 homologue A
172887
41
5



that interacts with Disco


O60673
DNA polymerase zeta
0
32
3



catalytic subunit


Q02880
Topoisomerase 2-beta DNA
184682
33
3


P27695
(apurinic or apirimidinic
35979
50
2



site) DNA lyase


O14802
RPC1 subunit of RNA polymerase
0
36
2



III directed at DNA


O75165
Member 13 of DnaJ homolog
0
35
3



subfamily


Q6PKX4
Coupling protein 6
0
42
5


Q8IVF4
10 dyneine heavy chain,
0
48
4



axonemal


Q96DT5
11 dyneine heavy chain,
0
38
7



axonemal


Q9UFH2
Heavy chain 17 of dynein,
517327
42
5



axonemal


Q9P225
2 dyneine heavy chain,
0
37
6



axonemal


Q8TD57
Heavy chain 3 dynein,
476111
48
5



axonemal


Q8TE73
5 dyneine heavy chain,
0
42
5



axonemal


Q96M86
Protein 1 containing
540950
44
4



dinein heavy chain domain


Q03001
Dystonia
867875
62
10


Q7Z6Z7
E3 ubiquitin-protein
0
51
4



ligase HUWE1


Q6ZT12
E3 ubiquitin-protein
0
33
2



ligase UBR3


Q8IUD2
CAST family member
0
37
3



1/interacting with



ELKS/Rab6


P68104
Elongation factor-alpha 1
50649
450
1


Q05639
Elongation factor-alpha 2
50962
173
1


P29692
1-delta elongation factor
31281
88
3



1


P26641
Gamma elongation factor 1
50525
201
1


P13639
Elongation Phantom 2
96711
350
5


O15083
ERC protein 2
0
44
4


P60842
Eukaryotic initiation
46628
152
6



factor 4A-I


P63241
Eukaryotic translation
17145
89
1



initiation factor 5A-I


Q86XX4
FRAS1 extracellular
454911
48
7



matrix protein


P15311
Ezrin
69726
450
13


P14324
Farnesyl pyrophosphate
48886
72
2



synthase


Q16658
Fascin
55198
348
7


P49327
Fatty acid synthase
276610
500
21


A0AVI2
Fer-1-similar to protein
0
36
3



5


Q4ZHG4
Protein 1 containing
206145
40
5



fibronectin type III



domain


O75369
Filamin-B
280749
41
4


Q68DA7
Formin-1
159015
46
5


Q5SZK8
FRAS1-related
0
33
5



extracellular matrix



protein 2


P04075
Fructose bisphosphate
39915
507
13



aldolase A


P09972
Fructose Bisphosphate
39894
181
3



Aldolase C


Q9UKJ3
Protein 8 containing
165218
39
4



patch domain G


P09382
Galectin-1
15080
43
2


Q08380
Galectin-3 binding
66361
154
5



protein


P09104
Gamma-enolase
47715
245
3


Q92820
Gamma-glutamyl hydrolase
36452
49
2


Q3V6T2
Girdin
0
49
2


P14136
Glial fibrillar acidic
50099
88
2



protein


P11413
Glucose-6-phosphate 1-
59907
68
3



dehydrogenase


P06744
Glucose-6-phosphate
63579
660
2



isomerase


Q8TCU5
Glutamate receptor 3A
127005
32
6



subunit [NMDA]


P78417
Glutathione S-transferase
27945
68
3



omega-1


P04406
Glyceraldehyde-3-phosphate
36361
619
3



dehydrogenase


P62826
GTP-binding nuclear Ran
24643
227
6



protein


Q8NDA8
Protein 7A containing HEAT
0
34
3



repeat


P0DMV8/
70 kDa 1A/1B protein from
70443
410
9


P0DMV9
thermal shock


P34931
Protein 1 of 70 kDa thermal
70913
381
8



shock-similar


P34932
Protein 4 of 70 kDa from
95525
76
3



thermal shock


P17066
Protein 6 of 70 kDa from
71608
320
5



thermal shock


P11142
71 kDa cognate thermal
71294
704
12



shock protein


Q12931
75 kDa protein from thermal
80654
108
2



shock, mitochondrial


P04792
Beta-1 thermal shock
22858
95
2



protein


P07900
HSP 90-alpha thermal shock
85333
367
11



protein


P08238
HSP 90-beta thermal shock
0
518
3



protein


P54652
70 kDa protein related
70428
398
7



thermal shock


Q96RW7
Hemicentin-1
624433
47
6


Q8NDA2
Hemicentin-2
550521
53
4


P09651
Heterogeneous nuclear
38933
117
3



ribonucleoprotein A1


Q32P51
Heterogeneous nuclear-
34471
117
3



similar A1



ribonucleoprotein 2


P61978
Heterogeneous nuclear
51426
117
3



ribonucleoprotein K


P22626
Heterogeneous nuclear
37576
122
4



A2/B1 ribonucleoproteins


Q9UQL6
Histone deacetylase 5
0
50
2


P0C0S8
Histone H2A type 1
14099
103
1


P04908
Histone H2A type 1-B/E
14143
103
1


Q93077
Histone H2A type 1-C
14113
103
1


P20671
Histone H2A type 1-D
14115
103
1


Q96KK5
Histone H2A type 1-H
13914
103
1


Q99878
Histone H2A type 1-J
13944
103
1


Q6FI13
Histone H2A type 2-A
14119
103
1


Q16777
Histone H2A type 2-C
14012
103
1


Q7L7L0
Histone H2A type 3
14129
103
1


Q9BTM1
Histone H2A.J
14027
103
1


Q71UI9
Histone H2A.V
13517
55
1


P0C0S5
Histone H2A.Z
13561
55
1


Q96A08
Histone H2B type 1-A
14207
41
2


P33778
Histone H2B type 1-B
13990
110
2


P62807
Histone H2B type 1-
13946
148
2



C/E/F/G/I


P58876
Histone H2B type 1-D
13976
148
2


Q93079
Histone H2B type 1-H
13932
148
2


P06899
Histone H2B type 1-J
13944
110
2


O60814
Histone H2B type 1-K
13930
148
2


Q99880
Histone H2B type 1-L
13992
148
2


Q99879
Histone H2B type 1-M
14029
148
2


Q99877
Histone H2B type 1-N
13962
148
2


P23527
Histone H2B type 1-O
13946
110
2


Q16778
Histone H2B type 2-E
13960
110
2


Q5QNW6
Histone H2B type 2-F
13960
148
2


Q8N257
Histone H2B type 3-B
13948
156
2


P57053
Histone H2B type F-S
13984
148
2


P68431
Histone H3.1
15557
64
3


Q16695
Histone H3.1t
15677
64
3


Q71DI3
Histone H3.2
15484
64
3


P84243
Histone H3.3
15408
66
3


Q6NXT2
Histone H3.3C
15350
66
3


P62805
Histone H4
11392
155
4


O14686
Histone-lysine N-
601158
69
6



methyltransferase MLL2


Q96L73
Histone-lysine N-
0
33
5



methyltransferase, H3



lysine-36 and H4 lysine-20



specific


Q4G0P3
Protein homolog that
582477
48
3



induces hydrocephalus


P00492
Hypoxanthine-guanine
24904
49
1



phosphoribosyltransferase


Q14974
Importin beta-1 subunit
98826
68
4


O00410
Importin-5
125730
34
2


Q14573
Inositol 1,4,5-trisphosphate
0
35
6



receptor type 3


P13645
Keratin, type I
59120
1084
11



cytoskeletal 10


Q99456
Keratin, type I
53832
101
3



cytoskeletal 12


P13646
Keratin, type I
50113
119
4



cytoskeletal 13


P02533
Keratin, type I
52117
113
3



cytoskeletal 14


P19012
Keratin, type I
49653
99
3



cytoskeletal 15


P08779
Keratin, type I
51754
261
3



cytoskeletal 16


Q04695
Keratin, type I
48521
83
3



cytoskeletal 17


P08727
Keratin, type I
44223
74
2



cytoskeletal 19


Q2M2I5
Keratin, type I
55751
58
3



cytoskeletal 24


Q7Z3Z0
Keratin, type I
49970
42
2



cytoskeletal 25


Q7Z3Y8
Keratin, type I
50515
65
3



cytoskeletal 27


Q7Z3Y7
Keratin, type I
51318
45
2



cytoskeletal 28


P35527
Keratin, type I
62435
1034
14



cytoskeletal 9


Q9NSB2
Keratin, type II cuticular
66102
71
2



Hb4


P04264
Keratin, type II
66301
1422
21



cytoskeletal 1


Q7Z794
Keratin, type II
62329
155
7



cytoskeletal 1b


P35908
Keratin, type II epidermal
65795
941
14



cytoskeletal 2


Q01546
Keratin, type II oral
66588
109
3



cytoskeletal 2


P12035
Keratin, type II
64741
101
3



cytoskeletal 3


P19013
Keratin, type II
57816
177
3



cytoskeletal 4


P13647
Keratin, type II
62776
258
3



cytoskeletal 5


P02538
Keratin, type II
60421
348
6



cytoskeletal 6A


P04259
Keratin, type II
60448
346
8



cytoskeletal 6B


P48668
Keratin, type II
60401
341
8



cytoskeletal 6C


P08729
Keratin, type II
51572
118
3



cytoskeletal 7


Q3SY84
Keratin, type II
0
89
3



cytoskeletal 71


Q14CN4
Keratin, type II
56640
72
3



cytoskeletal 72


Q86Y46
Keratin, type II
59611
151
5



cytoskeletal 73


Q7RTS7
Keratin, type II
0
83
3



cytoskeletal 74


O95678
Keratin, type II
59942
185
5



cytoskeletal 75


Q8N1N4
Keratin, type II
0
38
2



cytoskeletal 78


Q5XKE5
Keratin, type II
58229
97
4



cytoskeletal 79


P05787
Keratin, type II
53927
159
4



cytoskeletal 8


Q02241
Kinesin-like protein KIF23
0
32
3


Q9ULI4
Kinesin-like protein
0
47
5



KIF26A


Q63ZY3
Protein 2 containing
0
42
2



repeat motif of KN motif



and anchyrine


Q03252
Lamin-B2
67985
33
2


P25391
Laminin alpha-1 subunit
0
35
5


P24043
Laminin alpha-2 subunit
353874
34
5


P48634
BAT2 protein rich in long
229564
48
4



proline


Q96JM7
Lethal malign brain tumor
89921
37
3



(3) - similar to protein 3


Q5S007
Repeating serine rich in
290688
49
4



leucine/threonine-protein



kinase 2


Q5VZK9
16A protein containing
153228
36
3



leucine-rich repetition


Q9UIQ6
Leucyl-cystinyl
0
31
2



aminopeptidase


Q8N3X6
Co-repressor of protein-
0
37
2



similar ligand-dependent



nuclear receptor


P00338
L-lactate dehydrogenase A
0
537
2



chain


Q9BYZ2
L-lactate dehydrogenase A-
42508
147
2



similar to 6B


P07195
L-lactate dehydrogenase B
37065
673
1



chain


Q9NZR2
1B protein related to low
536259
34
4



density lipoprotein



receptor


O75096
Protein 4 related to low
217797
50
6



density lipoprotein



receptor


Q12912
Lymphoid restricted
0
35
2



membrane protein


P10619
Lysosomal Protein
55145
41
1


P40925
Malate dehydrogenase,
36775
168
2



cytoplasmic


Q8IWI9
Gene MAX associated
0
41
3



protein


Q15648
RNA polymerase II
0
46
3



transcription subunit



mediator 1


Q96JG8
Melanoma associated D4
81843
41
2



antigen


Q13421
Mesothelin
69770
41
2


P01033
Metalloproteinase
23904
61
2



inhibitor


Q96PK2
Factor 1 crosslinking
675712
58
9



microtubule-actin, isoform 4


Q9UPN3
Microtubule-actin
0
42
6



crosslinking factor 1,



1/2/3/5 isoforms


P78559
Protein 1A associated with
0
35
4



microtubule


P11137
Microtubule associated
0
37
4



protein 2


O60307
Serine associated with
144213
44
3



microtubule/threonine-



protein kinase 3


Q5JR59
Candidate 2 tumor
151203
33
2



suppressor associated with



microtubule


Q9NU22
Midasin
640088
42
7


Q8N4C8
Deformation-similar to
150873
45
2



kinase 1


Q99797
Mitochondrial peptidase
81817
34
5



intermediary


O95819
Protein kinase 4 activated
0
37
2



by mitogen


P26038
Moesin
68198
466
12


Q8WXI7
Mucin-16
2366624
80
9


Q7Z5P9
Mucin-19
0
37
4


Q15746
Myosin light chain kinase,
213816
44
3



smooth muscle


P35749
Miosin-11
0
42
3


Q7Z406
Miosin-14
229353
33
4


A7E2Y1
Miosin-7B
0
41
5


P13535
Miosin-8
224298
33
4


P35579
Miosin-9
228467
51
3


Q9UKN7
Miosin-XV
398694
35
3


Q92614
Miosin-XVIIIa
234920
55
6


Q6T4R5
Nance-Horan syndrome
178183
33
4



protein


Q86VF7
Nebulin-related anchor
198688
43
3



protein


Q8WXH0
Nesprin-2
0
48
6


Q6ZNJ1
Neurobeachin-similar to
306093
44
3



protein 2


Q09666
AHNAK protein associated with
633316
77
7



neuroblast differentiation


P21359
Neurofibromin
0
33
5


Q8NEY1
Neuron Browser 1
203760
90
7


Q15818
Neural Pentraxin-1
47727
119
4


Q99574
Neuroserpin
46717
103
1


Q86XR2
Niban-like protein 2
0
39
4


P29474
Nitric oxide synthase,
135130
36
4



endothelial


Q15233
Octamer binding protein
0
51
1



containing non-POU domain


Q8IVI9
Nostrin
0
42
3


Q92621
Complex pore nuclear Nup25
0
38
3



protein


P15531
Nucleoside diphosphate
17389
354
2



kinase A


P22392
Nucleoside diphosphate
17481
273
8



kinase B


Q9NTK5
Obg-similar to ATPase 1
45059
100
1


Q5VST9
Obscurin
881261
56
5


Q504Q3
PAB-dependent poly (A)
137244
37
3



specific ribonuclease



subunit 2


Q9BYG4
Gamma counterpart of
41150
34
2



defective partition 6


P62937
Peptidyl-prolyl cis-trans
18309
225
3



isomerase A


P23284
Peptidyl-prolyl cis-trans
23865
319
6



isomerase B


Q06830
Peroxiredoxin-1
22372
332
4


P32119
Peroxiredoxin-2
22065
296
6


Q13162
Peroxiredoxin-4
30765
202
5


P30041
Peroxiredoxin-6
25197
379
13


P30086
Phosphatidylethanolamine
21190
73
1



binding protein 1


P36871
Phosphoglucomutase-1
61892
230
8


P00558
Phosphoglycerate kinase 1
45216
710
18


P07205
Phosphoglycerate kinase 2
45413
181
7


P18669
Phosphoglycerate mutase 1
28996
331
7


P13797
Plastin-3
71560
83
3


Q15149
Plectin
534517
58
8


P11940
Polyadenylate Binding
71306
51
5



Protein 1


Q8TDX9
Polycystic liver disease
0
44
4



protein-similar to 1


P98161
Polycystin-1
468373
87
7


Q6S8J3
Member E of the Anarchine
123582
307
12



POTE domain family


A5A3E0
F member of anchyrine POTE
123692
35
3



domain family


P0CG38
Member I of the Anchirine
123530
70
4



POTE family


O60809
Member 10 of the PRAME family
0
37
4


P02545
Prelamin-A/C
74540
81
3


P07602
proactivator polypeptide
60202
63
1


Q9HD20
ATPase 13A1 carrying
0
47
1



likely cation


Q9Y4D8
Likely E3 ubiquitin-
446102
48
4



protein ligase C12orf51


O75592
Likely E3 ubiquitin-
519504
48
4



protein ligase MYCBP2


Q9NR48
Likely histone-lysine N-
0
33
4



methyltransferase ASH1L


Q02809
Procollagen-lysine, 2-
84285
68
4



oxoglutarate 5-dioxygenase



1


P07737
Profilin-1
15296
306
6


P12004
Nuclear Cellular Antigen
29252
97
2



and Proliferation


Q9UQ80
2G4 protein associated
44299
144
1



with proliferation


Q8NBP7
Proprotein convertase
75866
423
2



subtilisin/kexin type 9


P25786
Alpha subunit type -1 of
29950
54
3



proteasome


P25787
Alpha subunit type -2
26076
140
4



proteasome


P25788
Proteasome type-3 alpha
28787
43
3



subunit


P25789
Proteasome type-4 alpha
29846
36
1



subunit


P60900
Alpha subunit type -6
27934
133
4



proteasome


O14818
Proteasome type-7 alpha
28089
130
5



subunit


P20618
Proteasome type-1 beta
26844
70
2



subunit


P28070
Proteasome type-4 beta
29450
94
4



subunit


P28074
Proteasome type-5 beta
28761
249
6



subunit


P28072
Proteasome type-6 beta
25713
143
4



subunit


Q8IVF2
AHNAK2 protein
620521
66
4


Q9UPA5
Bassoon protein
0
52
6


P07237
Nucleoside diphosphate
57567
177
7



kinase B


P30101
Obg-similar to ATPase 1
57281
269
10


Q15084
A6 disulfide isomerase
48577
161
4



protein


Q9Y6V0
Piccolo protein
0
36
6


Q14160
Protein counterpart to
175955
36
4



write


O95785
Wiz protein
180520
44
6


Q9NYQ8
Protocadherina Fat 2
483217
45
6


Q6V0I7
Protocadherina Fat 4
0
44
5


Q96JQ0
Protocadherin-16
0
34
4


P00491
Purine nucleoside
32517
210
7



phosphorylase


Q5VTE0
Putative elongation factor
50709
450
1



1-alpha-similar to 3


Q58FF8
HSP 90-beta 2 protein from
44671
83
5



putative thermal shock


P0C7M2
Putative heterogeneous
34469
117
3



nuclear A1-like



ribonucleoprotein-3


B8ZZ34
Putative shisa-8 protein
52202
48
4


P46087
Putative ribosomal RNA
0
44
3



NOP2 methyltransferase


Q9H853
Putative tubulin-similar
27915
77
1



to alpha-4B protein


P14618
M1/M2 isozymes of pyruvate
58736
742
2



kinase


P50395
Rab GDP beta dissociation
51287
318
7



inhibitor


Q13671
Ras and Rab Interactor
0
37
3


P51157
Ras-related Rab-28 protein
0
50
2


P10586
Tyrosine-protein
214774
35
5



phosphatase F receptor-



type


Q9P227
Rho GTPase activation
163666
38
2



protein 23


P60891
Ribose phosphate
35469
34
1



pyrophosphokinase 1


Q9H7B2
Homolog of factor 2 of
35907
47
3



ribosome production


Q9P2E9
Ribosome binding protein 1
153020
64
3


Q8N1G1
RNA exonuclease 1 homolog
132891
53
3


Q5TZA2
Rootletin
229066
53
5


Q92736
Rhinodine receptor 2
571865
36
7


Q15413
Ryanodine receptor 3
560001
40
3


Q9UQ35
Serine/arginine repetitive
300720
67
4



matrix protein 2


Q9Y3S1
Serine/threonine-protein
244868
47
3



kinase WNK2


P62140
Catalytic subunit of
38073
59
3



serine/threonine-protein



phosphatase PP1-beta


Q13315
ATM serine protein kinase
356828
37
5


P02787
Serotransferrin
79494
35
2


Q8IW75
Serpin A12
0
35
5


P35237
Serpin B6
43256
112
4


P50454
Serpin H1
46751
115
4


P02768
Serum albumin
71464
231
3


Q9UPX8
Protein 2 of SH3 and
0
59
3



multiple ankyrine repeat



domains


Q9BYB0
Protein 3 of SH3 and
0
57
4



multiple ankyrine repeat



domains


Q9H2Y9
Member 5A1 of solute
0
35
3



family carrier of organic



anion carrier


Q9H3E2
Classifying nexin-25
0
35
3


Q9UBP0
Spastin
67721
38
4


Q13813
Spectrin alpha chain,
285976
51
4



brain


Q9H254
Spectrin beta chain, brain
290693
86
4



3


Q9NRC6
Spectrin beta chain, brain
0
45
6



4


P11277
Spectrin beta chain,
0
46
4



erythrocyte


Q9P0W8
Protein 7 associated with
68430
64
3



spermatogenesis


Q13838
Spliceosome RNA helicase
49664
66
1



BAT1


Q12770
Sterol regulatory element
141819
48
2



binding protein cleavage



activation protein


Q15772
Striated muscle
357376
40
4



preferentially expressed



by protein kinase


O60279
Protein 5 containing Sushi
68953
37
1



domain


Q8TER0
Protein 1 containing
158397
53
5



domain similar to Sushi,



nidogen and EGF


Q9Y490
Talin-1
272726
40
4


Q9Y4G6
Talin-2
274829
56
4


Q5TCY1
Tau-tubulin kinase 1
0
33
1


P78371
Beta subunit of protein 1
58040
35
1



T complex


P50991
Delta subunit of protein 1
58650
77
2



T complex


P48643
Epsilon subunit of protein
60481
93
3



1 complex T


Q99832
Eta subunit of complex T
60107
71
1



protein


P49368
Gamma subunit of protein 1
61427
231
5



T complex


P50990
Theta subunit of protein 1
60450
70
5



T complex


P40227
Zeta subunit of protein 1
58676
141
5



T complex


Q99973
Component 1 of telomerase
0
41
4



protein


Q9UKZ4
Teneurin-1
0
36
6


Q9HBL0
Tensin-1
187026
52
3


Q5SRH9
39A tetratricpeptide
0
32
3



repeat protein


P10599
Thioredoxin
12063
40
2


Q16881
Thioredoxin reductase 1,
71841
36
4



cytoplasmic


P07202
Thyroid peroxidase
104851
65
6


Q8WZ42
Titin
3849990
140
36


P31629
HIVEP2 transcription
0
44
5



factor


P02786
Protein 1 transferrin
85506
115
4



receptor


Q15582
Ig-h3 protein
75496
52
1



transformation induced by



beta growth factor


P55072
Transitional endoplasmic
90282
449
11



ATPase reticulum


P29401
Transketolase
68739
742
1


P60174
Isomerase triosephosphate
26986
441
3


P07477
Tripsin-1
27159
44
2


P68363
Alpha-1B tubulin chain
50964
432
1


Q9BQE3
Alpha-1C tubulin chain
50708
430
1


P68366
Alpha-4A tubulin chain
50810
238
1


Q9NY65
Alpha-8 tubulin chain
50906
138
1


P07437
Beta tubulin chain
50375
400
1


Q9H4B7
Beta-1 tubulin chain
51147
103
2


P68371
Beta-2C tubulin chain
50551
54
1


P42684
Tyrosine protein kinase
129675
54
5



ABL2


P22314
Enzyme that activates
119260
163
10



ubiquitin-like modifier


Q5TEA3
C20orf194 protein without
134101
44
4



characterization


Q9Y4B5
KIAA0802 protein without
0
37
4



characterization


Q9Y2F5
KIAA0947 protein without
0
49
5



characterization


Q2LD37
KIAA1109 protein without
561392
47
6



characterization


Q5VZ46
KIAA1614 protein without
127754
35
3



characterization


O75445
Userin
587541
47
5


P46939
Utrophin
0
37
7


Q709C8
13C protein associated
0
62
4



with vacuolar protein



classification


Q96QK1
Protein 35 associated with
92765
88
2



classification of vacuolar



protein


P18206
Vinculin
0
56
3


O43497
Alpha-1G subunit of
266470
34
5



voltage-dependent type T



calcium channel


O75191
Xylulose kinase
0
34
4


Q96JG9
Zinc finger 469 protein
414696
43
7


Q9Y493
Zonadhesin
0
36
3









STM3 Interacts with HPV18 E7


Because in Mileo, A. M., Abbruzzese, C., Mattarocci, S., Bellacchio, E., Pisano, P., Federico, A., . . . Paggi, M. G. (2009). Human Papillomavirus-16 E7 Interacts with Glutathione S-Transferase P1 and Enhances Its Role in Cell Survival. PLoS ONE, 4 (10) is shown that GSTP1 protein interacts with HPV16 E7 protein and this interaction improves cell survival, to know if GSTM3 can interact with HPV 18 E7, an alignment was performed of structural overlap between the GSTP1 and GSTM3 proteins and the E7 proteins of HPV 16 and 18 using the MAMMOTH program, followed by the Swiss PDB Viewer (Deep View) v4.1 software to visualize the results (see FIG. 4A). The alignment showed conserved and unconserved regions by comparing the distances between alpha carbons and the main amino acid chain sequences.


To demonstrate this interaction, a vector construct was generated to express a GSTM3 recombinant human protein with a histidine tag added at the N-terminal (N-6× His-tag) in S. cerevisiae (see FIG. 4I, Table 6). GSTM3 was identified through anti-His western staining and peptide mass fingerprint analysis (see FIG. 4B, FIG. 4I). After capturing the GSTM3 recombinant protein, it was incubated with a HeLa cell protein extract (HPV18-positive) (see FIG. 4J). HPV18 E7 protein co-eluted with GSTM3 N-6λ-his-tag and was identified using a specific antibody by western spotting (see FIG. 4B). To verify this interaction, an HPV18 E7 construction in S. cerevisiae was generated, but it was possible to obtain a stable strain that expressed the protein. A plasmid construct was then generated that expressed an HPV18 E7 C-6λ-his-tag recombinant protein in the HeLa cell line and performed a protein interaction (“pull-down”) assay (see FIG. 4K). The results showed that GSTM3 can interact with HPV18 E7 (see FIG. 4C, FIG. 4L) and that this interaction acts similarly to the interaction between GSTP1 and E7 proteins of HPV16 (see FIG. 4A).









TABLE 6







List of primers used to generate


recombinant GSTM3 protein.








Name
Sequence 5′ > 3′





M3-1
ATGTCGTGCGAGTCGTCTATGGTTCTCGGGTACTGGGATATTC



GTGGGCTGGCGCACGCCATCCGCCTGCTCCTGG





M3-2
CATAGTCAGGAGCTTCCCCGCACGTGTACCGTTTCTCCTCATA



AGAGGTATCCGTGAACTCCAGGAGCAGGCGGATGG





M3-3
GGAAGCTCCTGACTATGATCGAAGCCAATGGCTGGATGTGAAA



TTCAAGCTAGACCTGGACTTTCCTAATCTGCCCTACC





M3-4
GCTTGCGAGCGATGTAGCGCAAGATGGCATTGCTCTGGGTGAT



CTTGTTCTTCCCATCCAGGAGGTAGGGCAGATTAGG





M3-5
GCTACATCGCTCGCAAGCACAACATGTGTGGTGAGACTGAAGA



AGAAAAGATTCGAGTGGACATCATAGAGAACC





M3-6
CAGTTTTTCGTGGTCAGAGCTGTAACAGAGCCTTATCAGTTGT



GTGCGGAAATCCATTACTTGGTTCTCTATGATGTCC





M3-7
GCTCTGACCACGAAAAACTGAAGCCTCAGTACTTGGAAGAGCT



ACCTGGACAACTGAAACAATTCTCCATGTTTCTGG





M3-8
GGTGAGAAAATCCACAAAGGTGAGCTTTTCCCCGGCAAACCAT



GAGAATTTCCCCAGAAACATGGAGAATTG





M3-9
CCTTTGTGGATTTTCTCACCTATGATATCTTGGATCAGAACCG



TATATTTGACCCCAAGTGCCTGGATGAGTTCC





M3-10
CCAAGTGCCTGGATGAGTTCCCAAACCTGAAGGCTTTCATGTG



CCGTTTTGAGGCTTTGGAGAAAATCGCTGCC





M3-11
CCACTGGGCCATCTTGTTGTTGATGGGCATCTTGCAGAACTGA



TCAGACTGTAAGTAGGCAGCGATTTTCTCCAAAGC





M3-12
CCATCAACAACAAGATGGCCCAGTGGGGCAACAAGCCTATATG



CTGA





GSTM3-
ATAGAC AAGCTT AACAAAATGTCTGGGTCGTCG


HindIII

CACCATCACCACCATCAT TCGTGCGAGTCGTCTATGG






GSTM3-
ATACAA GGATCC TCAGCATATAGGCTTGTTGC


BamHI









GSTM3-HindIII. This oligonucleotide contains the HindIII restriction site (in bold), yeast consensus sequence at the translational start site and codons for 6 histidines (bold and underlined).


GSTM3-BamHI. This oligonucleotide contains the BamHI restriction site (bold).


Primers used for amplification and cloning of the HPV18 E7 gene











E718-1



ATG CAT GGA CCT AAG GCA ACC ATT







E718-2*



CTG CTG GGA TGC ACA CCA







E7-18-Hind III



ATA CAA AAG CTT ATG CAT GGA CCT AA







E7HPV18-his*




GAT GGT GAT GAT G CT GCT GG








Univ His-Tag BamH I*



TAC GTG GAT CCT AGT GGT GAT GGT G






E7-18-Hind III. This oligo contains the Hind III restriction site (bold) and initial HPV 18 sequence (bold).


E7HPV18-his. This oligo contains a fragment of 6× histidine sequence (bold).


Univ His-Tag BamHI. This oligo contains the BamHI restriction site (bold) and 6× histidine sequence fragment (bold and underlined).


Once the interaction of the GSTM3 protein with the HPV18 E7 protein was demonstrated, the relevance of this interaction in cell survival was evaluated. For this purpose, a stress sensitivity test with UV was developed in a breast cancer cell line MDA-MB-231 which is negative for HPV, and the expression of GSTM3 and GSTP1 proteins (see FIG. 4G). Using recombinant GST and E7 HPV18 proteins, a phenotype analysis was performed by exogenous protein complementation (PAEP). This analysis demonstrated that under stress of UV radiation (15 seconds UV, IC50), the GSTM3/HPV18 E7 cells exhibited a survival rate of 84.1%, while the GSTP1/GSTM3/E7 cells exhibited a survival rate of 93.7% after a 24-hour recovery period. These results indicate that there is a synergistic effect between GST and viral proteins (see FIG. 4D). An in vitro assay was performed wherein CC cell lines and negative cell lines were exposed to 6 mM cisplatin. This concentration of cisplatin completely killed the MDA-MB-231 cell line on the 4th day of treatment. For the HaCaT cell line, the total number of dead cells was on the 6th day (see FIG. 4E). Surprisingly, the cell lines that coexpress GST and HPV E7 survived for at least eight days after treatment (SiHa 17% and HeLa 24% confluence) (see FIG. 4E). To demonstrate that the GST/HPV18 E7 interaction was responsible for this resistance, a PAEP assay was performed using MDA-MB-231 cells that included recombinant proteins GSTM3, GSTP1 and E7 of HPV18. The results confirmed that cell lines expressing members of the HPV GST and E7 family of proteins have an advantage in terms of cell survival when treated with a xenobiotic agent (see FIGS. 4E, 4F). An increase in the survival of cells expressing any of these proteins (HPV18 E7, GSTM3 or GSTP1) was observed; however, the greatest increase in survival was observed when both GST and HPV18 E7 were present (see FIG. 4F).}


“Inhibition of Genes” (Knock-Down) GST In Vitro and In Vivo Using Antisense Oligonucleotides


As an example, without being limiting, the antisense oligonucleotides of vivo-morpholinos were designed to inhibit the therapeutic targets of the GSTs starting from the 5′UTR region of the messenger RNAs of the GSTM3 and GSTP1 and the ATG start codon and include 25 nucleotides, but this region is not limiting being able to use a range of 15-50 nucleotides, preferably from 18 to 30, more preferably from to 25, and bases 1-773, preferably close to the start codon, of the GSTP1 gene and for GSTM3 from base 1-4144, preferably close to the start codon, with a similarity of 100-50% of both sequences.


It is obvious that a skilled person in the art can employ any chemical modification of the RNA or DNA sequences to inhibit GSTs, for example: 2′MOE, 2′MO, PNA, LNA, Phosphorothioate, 2′-F, etc. They are commercially available. The preferred GSTs in the present invention, without being limiting thereof, are GSTM3 and GSTP1 with the sequence of antisense oligonucleotide (OAS) 5′-TAGACGACTCGCACGACATGGTGAC-3′ (56% CG−) and 5′-AATAGACCACGGTGTAGGGCG-3G′ (56% CG), respectively.


For both in vitro and in vivo tests both antisense oligonucleotides were dissolved in sterile PBS saline phosphate buffer at pH 7.5.


In order to evaluate the effect of GSTM3 and GSTP1 on CC cell lines, the expression of both proteins was inhibited by means of antisense oligonucleotides (OAS) and a random sequence was used as a control. To evaluate the inhibition of GSTs, eight doses were evaluated for each antisense oligonucleotide (OAS) in culture with two cell lines, HeLa and HaCaT (negative control). The concentrations used were between the range of 10 to 1,280 ng/mL and were incorporated into the culture medium. Subsequently, cell proliferation was evaluated at three different times at 24, 48 and 72 hours. It was observed that HaCaT cells were not affected by treatment with any antisense oligonucleotide during the analysis period. However, a slight loss of survival was noted with the highest dose (1,280 ng/mL). In HeLa cells, viability losses were observed after 48 hours of treatment in all doses of OAS-GSTM3 (see FIG. 5A). After 72 hours, the highest treatment doses (640 and 1,280 ng/mL) showed a survival of less than 10% compared to the control cells. Similar results were obtained for treatment with OAS-GSTP1 in both cells.


To evaluate the cellular response in other cell lines, the dose of 640 ng/mL was selected, because this is the highest dose that did not affect the HaCaT cell line. In addition, treatment was performed with 640 ng/mL in the SiHa CC cell line (see FIG. 5B). A very similar response was obtained between cancer cell lines, indicating that both GST proteins are essential for cell survival in CC, but not for HaCaT (non-cancerous) cells. To validate the effectiveness of the elimination treatment, a western blot analysis was performed on the three cell lines for both proteins (see FIGS. 5D-5E). Immunoblotting revealed that both proteins were in fact negatively regulated during all times of treatment in all three cell lines. In addition, the cell viability in the three cell lines was evaluated after 24 and 48 hours of treatment at the dose of 640 ng/mL of the two antisense oligonucleotides (see FIG. 5C). A live/dead cell assay was carried out based on the staining of Syto9/propidium iodide. The results confirmed that HaCaT cells were not affected by the treatment. Both cancer cells were similarly affected. Together, these results demonstrate that HaCaT cells have an alternative mechanism of cell maintenance that is compromised to CC cells.


For in vivo assays, 15 days after tumor inoculation in mice, six doses of 400 ng/500 μL were injected intratumorally, every third day. On day 30, tumors were collected for later analysis. A randomized antisense oligonucleotide was used as a control in both in vitro and in vivo assays. The sequences of the antisense oligonucleotides used in FIGS. 6A-6E.


Loss of GST Inhibits Tumor Progression in Cervical Cancer


To demonstrate the importance of GST during tumor progression (PT), the effects of treatments with antisense oligonucleotides in a murine model were examined (see FIG. 6A). For this, the antisense oligonucleotides (OAS-GSTM3, OAS-GSTP1, and OAS-Control) were used and four CC cell lines were treated (two HPV16-positive lines, SiHa and CaSki, and two HPV18-positive lines, HeLa and Calo), as well as two different cell lines to CC, one of breast cancer (MDA-MB-231) and one of colon (COLO 205). The results of the in vivo and in vitro analyzes were correlated with each other, showing a drastic decrease in the volume in the tumor cell lines of CC (see FIGS. 6B-6C). However, the results for HeLa tumors were different from those performed in vitro. HeLa tumors only expressed GSTM3, but not GSTP1 (see FIGS. 5D-5E). Therefore, treatment with OAS-GSTP1 in HeLa tumors did not affect PT, which confirmed that GSTP1 is not expressed in these tumors. On the other hand, treatment with OAS-GSTM3 in HeLa tumors dramatically decreased tumor volume. Compared to the treatment with the control antisense oligonucleotide, the tumor volume of HeLa with OAS-GSTM3 was 14 times lower (see FIGS. 6B-6E).


In CaLo tumors, both proteins expressed as GSTM3 and GSTP1 were found (see FIGS. 6D-6E). Treatment of these tumors with the antisense oligonucleotides against GSTM3 and GSTP1 resulted in a decrease in tumor volume 10 and 6 times, respectively (see FIGS. 6B-6C). In the case of SiHa tumors, which express both proteins (see FIGS. 6D-6E), it was observed that the greatest decreases in tumor volume after treatment with both antisense oligonucleotides were 43 and 62 fold decreases for OAS-GSTM3 and OAS-GSTP1, respectively (see FIGS. 6B-6C). CaSki cell line control tumors also expressed both proteins. In the treated tumors, it was observed that the levels of GSTM3 and GSTP1 did not decrease as much as in other tumors that expressed these proteins (see FIGS. 6D-6E). Treatment with OAS-GSTP-1 resulted in a tumor volume reduction of 2.6 times compared to the control. In the case of treatment with OAS-GSTM3, no significant differences were observed between the volume of control tumors and tumors treated with OAS-GSTM3 (see FIGS. 6B-6C). The low efficacy in the inhibition of protein expression by treatment, particularly for GSTM3, was responsible for the poor response in tumor reduction, so the remaining GSTM3 is sufficient to provide a protective effect to tumor cells.


In addition, the response to the treatment of tumors of two cell lines of different origins, MDA-MB-231 of breast cancer and COLO of colon cancer, was also studied. Both tumors exhibited low GSTP1 expression compared to CC tumors (see FIGS. 6D-6E). However, the treated COLO tumors had levels 1.9 times lower than the control (see FIGS. 6B-6C). In the case of MDA-MB-231, GSTP1 levels were barely detectable in control tumors and, as a consequence, treatment with OAS-GSTP1 did not affect PT (see FIGS. 6B-6C). Tumors of both cell lines (COLO and MDA) expressed GSTM3 and in both cases, treatment with the antisense oligonucleotide significantly reduced protein levels.


GSTM3 and GSTP1 Regulate the MAP Kinase Proteins pJNK and pp38.


The effects of the deactivation of GSTM3 and GSTP1 on the activation of pJNK and pp38 and the phosphorylation of p65 and pERK (from the NF-κB pathway) during CC PT were analyzed (see FIGS. 7A-7H). Protein expression was analyzed through immunohistochemical assays in all CC tumors treated with antisense oligonucleotides (OAS-GSTM3, OAS-GSTP1 and OAS-control). OAS-GST treatments resulted in phosphorylation and activation of pJNK and pp38 MAP kinases. HeLa tumors that only express GSTM3 and, therefore, only responded to treatment with the OAS-GSTM3 antisense oligonucleotide, showing increased phosphorylation of JNK and p38 (see FIGS. 7A-7B). On the other hand, the CaLo and SiHa tumors only showed p38 phosphorylation, with the two treatments OAS-GSTM3 and OAS-GSTP1; CaLo (see FIGS. 7C-7D); and SiHa (see FIGS. 7E-7F). For CaSki tumors, both MAPK were positively regulated after treatment with OAS-GST (see FIGS. 7G-7H).


GSTM3 and GSTP1 Regulate Cell Survival by Inactivating NF-κB and pERK


The inactivation of the ERK protein and p65 NF-κB was examined (see FIGS. 7A-7H). HeLa tumors treated with OAS-GSTM3 showed inactivation of both proteins (see FIGS. 7A-7B).


In CaLo tumors, only pERK was inactivated after treatment with any of the antisense oligonucleotides for GST (see FIGS. 7C-7D). For SiHa tumors, only NF-κB was inactivated by any of the treatments (see FIGS. 7E-7F). In CaSki tumors, both proteins were inactivated after any treatment (see FIGS. 7G-7H). Inhibition of GSTM3 and GSTP1 proteins induced apoptosis and decreased cell survival through the NFκB and MAP kinase pathways.


GST Expression Analysis in Tissue Samples from CC Patients


A follow-up study of 13 patients with CC who had undergone chemotherapy was performed (see FIGS. 8E-8F). Protein expression analyzes were performed for GSTM3 and GSTP1 using immunohistochemistry (IHC). In this study, the percentage of the region of interest (ROI) that was immunopositive was analyzed. Surprisingly, all patients expressed both proteins, but with great variability with respect to the percentage of ROI (see FIG. 8A, FIGS. 8E-8F). Patients were arbitrarily categorized into three groups based on the percentage of ROI: weak, moderate and high for GSTM3 and GSTP1 (see FIGS. 8B-8C). Next, an association analysis of GST expression and patient survival was performed and two groups were generated: weak-moderate for GSTM3 and moderate for GSTP1 (DM-M), and another group with moderate-high values for GSTM3 and high values for GSTP1 (MA-A) (see FIG. 8D). The results showed that the expression of GSTM3 and GSTP1 could significantly influence the survival of patients with CC. A clear correlation is observed between patient survival and GST protein expression. Patients who showed a weak to moderate expression (DM-M) showed a significantly higher survival rate than patients who exhibited a moderate to high GST expression (MA-A) (see FIG. 8D). The results are shown in Table 7 below.









TABLE 7







ROI of GST proteins of patients with CC






















Mean sum
Total




Mean sum
Total






Mean sum
without
Area
Mean


Mean sum
without
Area
Mean




of stain
stain
Cell
area


of stain
stain
Cell
area




(Area)
(Area)
Area
fraction
Classifi-

(Area)
(Area)
Area
fraction
Classifi-


Protein
ID
(μm2)
(μm2)
(μm2)
ROI %
cation
Protein
(μm2)
(μm2)
(μm2)
ROI %
cation






















GSTM3
502
311,243.79
735,599.42
1,046,843.21
29.73
High
GSTP1
814,269.62
442,021.41
1,256,291.03
64.8
High



300
36,571.91
527,141.79
563,713.70
6.49
Weak

536,105.53
566,210.48
1,102,316.01
48.6
Moder-














ate



698
67,007.39
177,911.74
244,919.13
27.36
High

511,678.95
488,372.19
1,000,051.14
51.2
High



324
113,134.55
580,128.64
693,263.20
16.32
Moder-

895,090.00
448,948.52
1,344,038.52
66.6
High








ate



345
450,109.91
838,099.43
1,288,209.34
34.94
High

577,972.52
686,381.85
1,264,354.38
45.7
Moder-














ate



944
126,711.35
655,983.41
782,694.77
16.19
Moder-

516,617.31
696,180.99
1,212,798.30
42.6
Moder-








ate





ate



535
253,528.44
564,860.42
818,388.86
30.98
High

869,484.57
380,402.05
1,249,886.62
69.6
High



241
236,100.04
639,148.15
875,248.19
26.98
High

232,216.69
761,975.89
994,192.58
23.4
Moder-














ate



209
22,949.68
484,613.05
507,562.73
4.52
Weak

265,209.56
355,823.84
621,033.40
42.7
Moder-














ate



592
39,824.78
830,366.61
870,191.39
4.58
Weak

221,299.66
490,337.19
711,636.84
31.1
Moder-














ate



440
200,506.09
985,348.59
1,185,854.68
16.91
Moder-

877,113.35
420,341.69
1,297,455.04
67.6
High








ate



552
113,462.72
617,323.87
730,786.59
15.53
Moder-

240,094.59
345,836.90
585,931.49
41.0
Moder-








ate





ate



537
72,269.62
680,970.07
753,239.69
9.59
Weak

37,216.89
708,046.98
745,263.87
5.0
Weak









The results obtained indicate that patients with cervical cancer expressed the GSTM3 and GSTP1 proteins, and that the expression of the tissue samples is related to survival. Therefore, it is concluded that the increase of these proteins is involved with the prognosis of the patient being unfavorable is overexpression.


The present invention provides evidence for the identification and inhibition of the expression of GSTM3 and GSTP1. In the results of the cultures, the cervical cancer cells were drastically affected by the blockade of both GST, while the HaCaT (non-cancerous) control cells were not affected by the inhibition of these proteins, whereby the GSTM3 and GSTP1 are crucial for the survival and proliferation of cancer cells in culture. In inoculated tumors, it was observed that in those CC cell lines expressing at least one of these proteins, the tumor volume decreased dramatically after treatment with antisense oligonucleotides.


The present invention demonstrates that inhibition of GSTM3 or GSTP1 activates the signaling of JNK and p38, which leads the cells to apoptosis and, therefore, decreases the tumor volume. On the other hand, it was observed that the inactivation of NF-κB and/or ERK after GST inhibition inhibits cell survival.


The present invention shows that there is a strong association of the overexpression of GSTM3 and GSTP1 proteins and the survival of patients. The results obtained in vitro and in vivo are consistent with the clinical data, since the survival of patients with CC was associated with high levels of GST protein (see FIG. 7C). These data also agreed with studies on bladder and colon cancer in which it was found that GSTM3 protein overexpression was associated with a reduced patient survival rate. Therefore, the present invention presents a mechanism by which, CC cells use GST proteins to prevent apoptosis and activate cell survival and proliferation. In addition, this response is affected by the inhibition of these proteins (see FIG. 9).


EXAMPLES

The following examples will allow us to understand the present invention even more, as well as to show the best method of carrying it out. It should be understood that said examples are illustrative of the present invention and not in any way limiting the scope thereof. References cited herein should be construed as being expressly incorporated therein.


It should also be understood that the methods and techniques of protein extraction, and immunodetection, as well as the protocols for sample preparation, preparative two-dimensional gel electrophoresis, image analysis and protein identification through MALDI mass spectrometry, which are not specifically described in the examples, are reported in the aforementioned literature and are known to those skilled in the art. For example, phenolic protein extraction is described in Hurkman, W. J., & Tanaka, C. K. (1986).



Solubilization of plant membrane proteins for analysis by two-dimensional gel electrophoresis. Plant physiology, 81(3), 802-806; Encarnación, S., Guzmán, Y., Dunn, M. F., Hernández, M., Vargas, M. del C., & Mora, J. (2003). Proteome analysis of aerobic and fermentative metabolism in Rhizobium etli CE3. In Proteomics (Vol 3, bll 1077-1085), Klose, J., & Kobalz, U. (1995). Two-dimensional electrophoresis of proteins: an updated protocol and implications for a functional analysis of the genome. Electrophoresis, 16(6), 1034-1059.


Tumor Generation


Female athymic nude mice 4-6 weeks old (BALB/c Nu/Nu) were used and subcutaneously injected with 10′ tumor cells in 500 μL of RPMI 1640 medium without FBS and collected in 30, 45 and 50 days. The tumors were measured using a Vernier caliper, and the tumor volume was obtained by calculating the volume of an ellipsoid as π/6 (L*W*H), wherein L: length, W: width and H.


Tumor Protein Extraction, Proteomic Analysis and Mass Spectrometry.


Tumor tissue samples were extracted by macerating them in liquid nitrogen and a cocktail of protease inhibitor and phosphatases, followed by sonication on ice, to then carry out the extraction of phenolic proteins by extraction with RIPA buffer. Subsequently, the protein expression analysis is performed using anti-GSTM3 and anti-GSTP1 antibodies and then visualized by immunodetection techniques such as: western staining (western blot), immunohistochemistry, ELISA, etc.


Extracellular Protein Extraction In Vitro and Ex Vivo.


Cell lines were cultured with RPMI 1640 advanced serum free up to 70-80% confluence. The medium was removed and rinsed three times with sterile saline: 0.9% NaCl (w/v). After washing, FBS-free RPMI 1640 medium without fresh red phenol (Gibco) was added and incubated for 20 h. Later, the medium was recovered and centrifuged at 2,000 g for 5 minutes. The supernatant was passed through a 0.22 μm pore size PVDF membrane (Millex, Millipore) and stored at −70° C. until later use. For extracellular proteins from xenograft tumors, HeLa and SiHa tumors were inoculated with 107 cells. After 30, 45 and 50 days after inoculation, the tumors were collected and washed 3 times with saline. The procedure followed to extract secreted proteins from tumors was performed as previously described for the cells in culture and the supernatant was stored at −70° C. until later use.


Identification of Secreted Proteins Through LC-MS/NS


The identification of secreted proteins in cell lines was separated in SDS-PAGE. The generated peptides were analyzed in a nanoLC-MS/MS system (Q-TOF Synapt G2 MS; Waters), the identification of the peptides and proteins was done through the MASCOT Distiller interface (Matrix Science), and the database Swiss-Prot and NCBI.


Signal Peptide Analysis


For the analysis of the signal peptide a bioinformatic program called SignalP 4.1 was used, which predicts the presence and location of signal peptide sites in amino acid sequences. The method predicts and identifies the export sites of the signal peptide based on physicochemical features and a combination of neural networks (NN) and hidden Markov models (HMM).


Coimmunoprecipitation and Immunostaining (Immunoblot)


The HeLa tumor was collected at 50 days and stored at 80° C. until use. After the tumor sample, they were macerated in liquid nitrogen and lysed with 500 μL RIPA buffer (10 mM Tris, 1 mM EDTA, 1% NP40, 0.1% sodium deoxycholate, 140 mM NaCl) and supplemented with inhibitors of protease and phosphatase (10 mM β-glycerophosphate, 10 mM Na3VO4, 10 mM sodium fluoride). Total cell lysates were centrifuged at 13,000 g for 5 min to settle the insoluble material. The lysates are incubated 2 hours with sepharose protein A, normalized for the total protein concentration (10 μg protein) using SDS-PAGE. Protein candidate antibodies (GSTM3 and TRAF6) were immunoprecipitated by incubating lysates with 6 μL antibody conjugated sepharose overnight at 4° C. The beads were washed 3 times with 500 μL lysis buffer. Co-immunoprecipitant proteins were resolved in 12% SDS-PAGE. GSTM3 and TRAF6 levels were detected by immunoblotting using previously described anti-antibodies.


Commercial antibodies to analyze Western staining (western blot) proteins were selected from anti-GSTM3 (Abcam, ab67530, 1:10,000), anti-GSTP1 (Abcam, ab53943, 1:10,000), anti-TLR4 (Biolegen, 312804, 1:10,000), anti-TRAF6 (Abcam, ab13853, 1:10,000), anti-NF-Kb P65 (SC-378, 1:1,000), anti IKB-α (SC-371, 1:1,000), anti-JNK (sc-1648, 1:1,000), antiERK (sc-94, 1:1,000), anti p38 (sc-535, 1:1,000), anti-NF-κB phospho p65 (sc-101752, 1:1,000), anti-phospho-JNK (sc-6254, 1:1,000), anti-phospho-ERK (sc-7383, 1:1,000), phospho-p38 (sc-7973, 1:1,000), anti-phospho-IKB-α (Cell signaling, 1:1,000), anti-HSP70 and HSP60 (Biolegen, 648005 and 681502, 1:10,000), HPV18 E7 (Abcam, ab38743, 1:1,000), anti-His tag antibody (Invitrogen, 372900, 1:5,000). The cells are lysed in a buffer containing 100 mM Tris (pH 8.6); 4% SDS; 100 mM DTT; a protease inhibitor cocktail. To ensure lysis, pulses are given with a sonicator for 1 second for DNA fragmentation. The proteins are electrophoresed in SDS-PAGE from 12% to 15% and transferred to nitrocellulose membranes using a semi-dry system. The already transferred membranes are blocked with 5% skim milk or bovine serum albumin in a Tris saline buffer containing Tween 20 (TBST) for 15 minutes at 4° C., washed three times in TBST. Subsequently, albumin or skim milk is removed and washed 3 times with TBST, then the membrane is incubated with the primary antibody (anti-GSTM3 or anti-GSTP1) at 4° C. overnight and tested with primary antibody diluted and incubated at 4° C. overnight. After removing the primary antibody the membranes were incubated with a secondary antibody conjugated to peroxidase for 2 h and then the membrane was revealed with a solution of Carbazol (27.2% Carbazol Stock, 72.6% acetate buffer, 0.2% H2O2), Carbazol Stock: N,N-Dimethylformamide≥98% and 3-Amino-9-ethylcarbazole (Sigma-Aldrich) in a 1:8 (w/v) ratio to generate a red/brown. Relative quantifications were performed with ImageJ software.


To analyze tissue samples by immunohistochemistry (IHC), the percentage of the region of interest (ROI) that is the region that gives positive expression of GSTs is analyzed. The medical records were reviewed, taking into account the patient's previous medical history. All cases were subjected to an immunohistochemical analysis using anti-GSTM3 (Abcam, ab67530, 1:1,000) and, anti-GSTP1 (Abcam, ab53943, 1:1,000). The paraffin blocks were sampled at a tissue thickness of 5 μm and produced in duplicate for each slide. The analysis was performed in an automated immunocontainer (Ventana Medical Systems). Three parts of the tumor were evaluated separately in each sample, as was the presence of staining in the tumor cells. In this study, we analyzed the percentage of the region of interest (ROI) stained by the antibodies, as estimated using the CellSens (Olympus) software. The samples were divided into two groups to assess the association of protein expression and patient survival: (W-M) consisting of a weak ROI for GSTM3 and moderate ROI for GSTP1; and (MH-H) which groups a moderate/high ROI for GSTM3 and high ROI for GSTP1. Kaplan-Meier survival curves were used for this analysis using XLSTAT, with the Greenwood IC and a significance level of 95%.


In order to assess the effect of GSTM3 and GSTP1 on CC cells, the expression of both proteins was inhibited by the use of antisense oligonucleotides (OAS). Three OAS were designed, two to specifically block proteins and one with a random sequence as a negative control (OAS-GSTM3, OAS-GSTP1 and OAS-Control). First, eight doses were evaluated for each OAS in culture with two cell lines, HeLa (cervical cancer) and HaCaT (negative cancer control). The doses used were from 10 to 1,280 ng/mL total incorporated into the culture medium. And subsequently, we evaluate cell proliferation at three different times at 24, 48 and 72 hours. In this experiment, it was observed that HaCaT cells were not affected by treatment with OAS-GSTM3 during the analysis period. Only a slight loss of survival was noted with the highest dose (1,280 ng/mL). In HeLa cells, viability losses were noted after 48 hours of treatment in all doses of OAS-GSTM3 (see FIG. 5A). After 72 hours, the highest treatment doses (640 and 1,280 ng/mL) showed a survival of less than 10% compared to the control cells. Similar results were obtained for treatment with OAS-GSTP1 in both cells.


Once the protein expression of the GSTM3 and/or GSTP1 in the tumor tissue is identified, the specific antisense oligonucleotides are administered (for example: 2′O-Me, 2′O-MOE, vivo-Morpholino, Morpholinos, LNA, PNA, among others) to perform the silencing of GSTs proteins, which will induce tumor cell death.


Although the present invention has been described with particularity in accordance with certain of the preferred embodiments, the examples should be interpreted only as illustrative of the invention and not for the purpose of limiting it. References cited herein are expressly incorporated for reference.


BIBLIOGRAPHY



  • Adler, V., Yin, Z., Fuchs, S. Y., Benezra, M., Rosario, L., Tew, K. D., . . . Ronai, Z. (1999).

  • Regulation of JNK signaling by GSTp. The EMBO journal, 18(5), 1321-1334.

  • Akira, S., & Takeda, K. (2004). Toll-like receptor signalling. Nature reviews. Immunology, 4(7), 499-511.

  • Asea, A., Rehli, M., Kabingu, E., Boch, J. A., Baré, O., Auron, P. E., . . . Calderwood, S. K. (2002). Novel signal transduction pathway utilized by extracellular HSP70. Role of toll-like receptor (TLR) and TLR4. Journal of Biological Chemistry, 277(17), 15028-15034.

  • Atkinson, A. J., Colburn, W. A., DeGruttola, V. G., DeMets, D. L., Downing, G. J., Hoth, D. F., . . . Zeger, S. L. (2001). Biomarkers and surrogate endpoints: Preferred definitions and conceptual framework. Clinical Pharmacology and Therapeutics. https://doi.org/10.1067/mcp.2001.113989 Beavis, A. L., Gravitt, P. E., & Rositch, A. F. (2017). Hysterectomy-corrected cervical cancer mortality rates reveal a larger racial disparity in the United States. Cancer, 123(6), 1044-1050.

  • Bild, A. H., Potti, A., & Nevins, J. R. (2006). Linking oncogenic pathways with therapeutic opportunities. Nature Reviews Cancer.

  • Bosch, F. X., Lorincz, a, Munoz, N., Meijer, C. J. L. M., & Shah, K. V. (2002). The causal relation between human papillomavirus and cervical cancer. Journal of Clinical Pathology, 55(4), 244-265.

  • Brekke, O. H., & Sandlie, I. (2003). Therapeutic antibodies for human diseases at the dawn of the twenty-first century. Nature Reviews Drug Discovery.

  • Brewer, C. A., Setterdahl, J. J., Li, M. J., Johnston, J. M., Mann, J. L., & McAsey, M. E. (2000). Endoglin expression as a measure of microvessel density in cervical cancer. Obstetrics and Gynecology, 96(2), 224-228.

  • Burk, R. D., Chen, Z., Saller, C., Tarvin, K., Carvalho, A. L., Scapulatempo-Neto, C., . . . Mutch, D. (2017). Integrated genomic and molecular characterization of cervical cancer. Nature, 543(7645), 378-384.

  • Cabelguenne, A., Loriot, M. A., Stucker, I., Blons, H., Koum-Besson, E., Brasnu, D., . . . De Waziers, I. (2001). Glutathione-associated enzymes in head and neck squamous cell carcinoma and response to cisplatin-based neoadjuvant chemotherapy. International Journal of Cancer, 93(5), 725-730.

  • Cekanova, M., & Rathore, K. (2014). Animal models and therapeutic molecular targets of cancer: Utility and limitations. Drug Design, Development and Therapy. https://doi.org/10.2147/DDDT.S49584

  • Chang, J., Jung, H. H., Yang, J. Y., Lee, S., Choi, J., Im, G. J., & Chae, S. W. (2014). Protective effect of metformin against cisplatin-induced ototoxicity in an auditory cell line. JARO—Journal of the Association for Research in Otolaryngology, 15(2), 149-158.

  • Cho, W. C. S. (2007). Contribution of oncoproteomics to cancer biomarker discovery. Molecular Cancer.

  • Dasari, S., Wudayagiri, R., & Valluru, L. (2015). Cervical cancer: Biomarkers for diagnosis and treatment. Clinica Chimica Acta.

  • Denny, L. E. (2008). Prevention of cervical cancer. Reproductive health matters, 16(32), 18-31.

  • Dueñas-Gonzalez, A., Lizano, M., Candelaria, M., Cetina, L., Arce, C., & Cervera, E. (2005). Epigenetics of cervical cancer. An overview and therapeutic perspectives. Mol Cancer, 4, 38. https://doi.org/1476-4598-4-38[pii]\n10.1186/1476-4598-4-38

  • Encarnación, S., Guzmán, Y., Dunn, M. F., Hernández, M., Vargas, M. del C., & Mora, J. (2003).

  • Proteome analysis of aerobic and fermentative metabolism in Rhizobium etli CE3. In Proteomics (Vol 3, bll 1077-1085).

  • Gillet, E., Meys, J. F. A., Verstraelen, H., Verhelst, R., De Sutter, P., Temmerman, M., & Broeck, D. Vanden. (2012). Association between Bacterial Vaginosis and Cervical Intraepithelial Neoplasia: Systematic Review and Meta-Analysis. PLoS ONE, 7(10), e45201.

  • Green, J. A., Kirwan, J. M., Tierney, J. F., Symonds, P., Fresco, L., Collingwood, M., & Williams, C. J. (2001). Survival and recurrence after concomitant chemotherapy and radiotherapy for cancer of the uterine cervix: a systematic review and meta-analysis. Lancet, 358(9284), 781-786.

  • Hamburg, M. A., & Collins, F. S. (2010). The Path to Personalized Medicine—Perspective. The New England Journal of Medicine, 363(4), 301-304. https://doi.org/10.1056/NEJMp1006304 Hanahan, D., & Weinberg, R. A. (2000). The hallmarks of cancer. Cell, 100(1), 57-70.

  • Hanash, S. M., Pitteri, S. J., & Faca, V. M. (2008). Mining the plasma proteome for cancer biomarkers. Nature.

  • Hanash, S., & Taguchi, A. (2010). The grand challenge to decipher the cancer proteome. Nat Rev Cancer, 10(9), 652-660.

  • Hayden, M. S., & Ghosh, S. (2012). NF-kB, the first quarter-century: Remarkable progress and outstanding questions. Genes and Development, 26(3), 203-234.

  • Hayes, J. D., Flanagan, J. U., & Jowsey, I. R. (2005). Glutathione transferases. Annu Rev Pharmacol Toxicol, 45, 51-88.

  • Holohan, C., Van Schaeybroeck, S., Longley, D. B., & Johnston, P. G. (2013). Cancer drug resistance: an evolving paradigm. Nature Reviews Cancer, 13(10), 714-726.

  • Hu, Z., Zhu, D., Wang, W., Li, W., Jia, W., Zeng, X., . . . Ma, D. (2015). Genome-wide profiling of HPV integration in cervical cancer identifies clustered genomic hot spots and a potential microhomology-mediated integration mechanism. Nature Genetics, 47(2), 158-163.

  • Huang, J., Tan, P.-H., Thiyagarajan, J., & Bay, B.-H. (2003). Prognostic significance of glutathione Stransferase-pi in invasive breast cancer. Modern pathology: an official journal of the United States and Canadian Academy of Pathology, Inc, 16(6), 558-565.

  • Hurkman, W. J., & Tanaka, C. K. (1986). Solubilization of plant membrane proteins for analysis by two-dimensional gel electrophoresis. Plant physiology, 81(3), 802-806.

  • IARC. (2016). Globocan 2012. Http://Www-Dep.Iarc.Fr/, GLOBOCAN, 2012-2013.

  • Jankova, L., Robertson, G., Chan, C., Tan, K. L., Kohonen-Corish, M., Fung, C. L.-S., . . . Clarke, S.

  • J. (2012). Glutathione S-transferase Pi expression predicts response to adjuvant chemotherapy for stage C colon cancer: a matched historical control study. BMC cancer, 12(1), 196.

  • Josic, D., Clifton, J. G., Kovac, S., & Hixson, D. C. (2008). Membrane proteins as diagnostic biomarkers and targets for new therapies. Current Opinion In Molecular Therapeutics, 10(2), 116-123. Opgehaal van.

  • Junod, S. W. (2013). FDA and Clinical Drug assays: A Short History. Opgehaal van.

  • Kawano, M., Mabuchi, S., Matsumoto, Y., Sasano, T., Takahashi, R., Kuroda, H., . . . Kimura, T. (2015). The significance of G-CSF expression and myeloid-derived suppressor cells in the chemoresistance of cervical cancer. Scientific Reports, 5(April), 1-13.

  • Klose, J., & Kobalz, U. (1995). Two-dimensional electrophoresis of proteins: an updated protocol and implications for a functional analysis of the genome. Electrophoresis, 16(6), 1034-1059.

  • Kogo, R., How, C., Chaudary, N., Bruce, J., Shi, W., Hill, R. P., . . . Liu, F.-F. (2014). The microRNA-218-Survivin axis regulates migration, invasion, and lymph node metastasis in cervical cancer. Oncotarget, 6(2), 1090-1100.

  • Kueng, W., Silber, E., & Eppenberger, U. (1989). Quantification of cells cultured on 96-well plates.

  • Analytical Biochemistry, 182(1), 16-19.

  • Laborde, E. (2010). Glutathione transferases as mediators of signaling pathways involved in cell proliferation and cell death. Cell Death and Differentiation.

  • Loktionov, a, Watson, M. a, Gunter, M., Stebbings, W. S., Speakman, C. T., & Bingham, S. a. (2001). Glutathione-S-transferase gene polymorphisms in colorectal cancer patients: interaction between GSTM1 and GSTM3 allele variants as a risk-modulating factor. Carcinogenesis, 22(7), 1053-1060.

  • Louie, S. M., Grossman, E. A., Crawford, L. A., Ding, L., Camarda, R., Huffman, T. R., . . . Nomura, D. K. (2016). GSTP1 Is a Driver of Triple-Negative Breast Cancer Cell Metabolism and Pathogenicity. Cell Chemical Biology, 23(5), 567-578.

  • Makridakis, M., & Vlahou, A. (2010). Secretome proteomics for discovery of cancer biomarkers. Journal of Proteomics.

  • Mao, C., Hughes, J. P., Kiviat, N., Kuypers, J., Lee, S. K., Adam, D. E., & Koutsky, L. A. (2003). Clinical findings among young women with genital human papillomavirus infection. American Journal of Obstetrics and Gynecology, 188(3), 677-684.

  • Matsuzawa, A., Saegusa, K., Noguchi, T., Sadamitsu, C., Nishitoh, H., Nagai, S., . . . Ichijo, H. (2005).

  • ROS-dependent activation of the TRAF6-ASK1-p38 pathway is selectively required for TLR4-mediated innate immunity. Nature immunology, 6(6), 587-592.

  • Medeiros, R., Vasconcelos, A., Costa, S. Lopes, C. (2004). Metabolic Susceptibility Genes and Prostate Cancer Risk in a Southern European Population: The Role of Glutathione S-Transferases GSTM1, GSTM3, and GSTT1 Genetic Polymorphisms. Prostate, 58(4), 414-420.

  • Meding, S., Balluff, B., Elsner, M., Schöne, C., Rauser, S., Nitsche, U., . . . Walch, A. (2012). Tissuebased proteomics reveals FXYD3, S100A11 and GSTM3 as novel markers for regional lymph node metastasis in colon cancer. Journal of Pathology, 228(4), 459-470.

  • Mileo, A. M., Abbruzzese, C., Mattarocci, S., Bellacchio, E., Pisano, P., Federico, A., . . . Paggi, M. G. (2009). Human Papillomavirus-16 E7 Interacts with Glutathione S-Transferase P1 and Enhances Its Role in Cell Survival. PLoS ONE, 4(10).

  • Mitra, A., MacIntyre, D. A., Marchesi, J. R., Lee, Y. S., Bennett, P. R., & Kyrgiou, M. (2016). The vaginal microbiota, human papillomavirus infection and cervical intraepithelial neoplasia: What do we know and where are we going next? Microbiome. https://doi.org/10.1186/s40168-016-0203-0

  • Mitra, A. P., Pagliarulo, V., Yang, D., Waldman, F. M., Datar, R. H., Skinner, D. G., . . . Cote, R. J. (2009). Generation of a concise gene panel for outcome prediction in urinary bladder cancer. Journal of Clinical Oncology, 27(24), 3929-3937.

  • Monaci, P., Luzzago, A., Santini, C., De Pra, A., Arcuri, M., Magistri, F., . . . Cortese, R. (2008).

  • Differential screening of phage-Ab libraries by oligonucleotide microarray technology. PLoS ONE, 3(1).

  • Moyer, V. A. (2012). Screening for Cervical Cancer: U.S. preventive services task force recommendation statement. Annals of Internal Medicine.

  • Mueller-Klieser, W. (1997). Three-dimensional cell cultures: from molecular mechanisms to clinical applications. Am J Physiol, 273(4 Pt 1), C1109-23.

  • Nishitoh, H., Saitoh, M., Mochida, Y., Takeda, K., Nakano, H., Rothe, M., . . . Ichijo, H. (1998). ASK1 is essential for JNK/SAPK activation by TRAF2. Molecular cell, 2(3), 389-395.

  • Offit, K. (2011). Personalized medicine: New genomics, old lessons. Human Genetics.

  • Ortiz, A. R., Strauss, C. E. M., & Olmea, O. (2002). MAMMOTH (matching molecular models obtained from theory): an automated method for model comparison. Protein science: a publication of the Protein Society, 11(11), 2606-2621.

  • Owen, L. A., Uehara, H., Cahoon, J., Huang, W., Simonis, J., & Ambati, B. K. (2012). Morpholino-mediated increase in soluble fit-1 expression results in decreased ocular and tumor neovascularization. PLoS ONE, 7(3).

  • Pavlou, M. P., & Diamandis, E. P. (2010). The cancer cell secretome: A good source for discovering biomarkers? Journal of Proteomics.

  • Pectasides, D., Kamposioras, K., Papaxoinis, G., & Pectasides, E. (2008). Chemotherapy for recurrent cervical cancer. Cancer Treatment Reviews.

  • Peralta-Zaragoza, O., Bermúdez-Morales, V. H., Pérez-Plasencia, C., Salazar-León, J., Gómez-Cerón, C., & Madrid-Marina, V. (2012). Targeted treatments for cervical cancer: A review. OncoTargets and Therapy.

  • Posadas, E. M., Simpkins, F., Liotta, L. a, MacDonald, C., & Kohn, E. C. (2005). Proteomic analysis for the early detection and rational treatment of cancer—realistic hope? Annals of oncology: official journal of the European Society for Medical Oncology/ESMO, 16(1), 16-22.

  • Price, J. C., Guan, S., Burlingame, A., Prusiner, S. B., & Ghaemmaghami, S. (2010). Analysis of proteome dynamics in the mouse brain. Proceedings of the National Academy of Sciences of the United States of America, 107(32), 14508-14513.

  • Quail, D. F., & Joyce, J. A. (2013). Microenvironmental regulation of tumor progression and metastasis. Nat Med, 19(11), 1423-1437.

  • Rose, P. G., Bundy, B. N., Watkins, E. B., Thigpen, J. T., Deppe, G., Maiman, M. A., . . . Insalaco, S. (1999). Concurrent cisplatin-based radiotherapy and chemotherapy for locally advanced cervical cancer. N Engl J Med, 340(15), 1144-1153.

  • Rouillard, A. D., Gundersen, G. W., Fernandez, N. F., Wang, Z., Monteiro, C. D., McDermott, M. G., & Ma'ayan, A. (2016). The harmonizome: a collection of processed datasets gathered to serve and mine knowledge about genes and proteins. Database, 2016, baw100.

  • Scarinci, I. C., Garcia, F. a R., Kobetz, E., Partridge, E. E., Brandt, H. M., Bell, M. C., . . . Castle, P. E. (2010). Cervical cancer prevention: New tools and old barriers. Cancer, 116(11), 2531-2542.

  • Scherf, U., Ross, D. T., Waltham, M., Smith, L. H., Lee, J. K., Tanabe, L., . . . Weinstein, J. N. (2000). A gene expression database for the molecular pharmacology of cancer. Nature genetics, 24(3), 236-244.

  • Scholle, F., Bendt, K. M., & Raab-Traub, N. (2000). Epstein-Barr Virus LMP2A Transforms Epithelial Cells, Inhibits Cell Differentiation, and Activates Akt. Journal of virology, 74 (22), 10681-10689.

  • Schwede, T., Kopp, J., Guex, N., & Peitsch, M. C. (2003). SWISS-MODEL: An automated protein homology-modeling server. Nucleic Acids Research, 31(13), 3381-3385.

  • Shannon, P., Markiel, A., Ozier, O., Baliga, N. S., Wang, J. T., Ramage, D., . . . Ideker, T. (2003). Cytoscape: A software Environment for integrated models of biomolecular interaction networks. Genome Research, 13(11), 2498-2504.

  • Siddik, Z. H. (2003). Cisplatin: mode of cytotoxic action and molecular basis of resistance.

  • Oncogene, 22(47), 7265-7279.

  • Sima, N., Wang, S., Wang, W., Kong, D., Xu, Q., Tian, X., . . . Ma, D. (2007). Antisense targeting human papillomavirus type 16 E6 and E7 genes contributes to apoptosis and senescence in SiHa cervical carcinoma cells. Gynecologic Oncology, 106(2), 299-304.

  • Simpson, R. J., Connolly, L. M., Eddes, J. S., Pereira, J. J., Moritz, R. L., & Reid, G. E. (2000). Proteomic analysis of the human colon carcinoma cell line (LIM 1215): development of a membrane protein database. Electrophoresis, 21, 1707-1732.

  • Tabas-Madrid, D., Nogales-Cadenas, R., & Pascual-Montano, A. (2012). GeneCodis3: A non-redundant and modular enrichment analysis tool for functional genomics. Nucleic Acids Research, 40(W1).

  • Tew, K. D., & Townsend, D. M. (2011). Regulatory functions of glutathione S-transferase P1-1 unrelated to detoxification. Drug metabolism reviews, 43(2), 179-193.

  • Tomayko, M. M., & Reynolds, C. P. (1989). Determination of subcutaneous tumor size in athymic (nude) mice. Cancer Chemotherapy and Pharmacology, 24(3), 148-154.

  • Townsend, D. M., & Tew, K. D. (2003). The role of glutathione-S-transferase in anti-cancer drug resistance. Oncogene, 22(47), 7369-7375.

  • Twombly, R. (2002). New Pap Test Terminology, Management Guidelines Published. Journal of the National Cancer Institute, 94(12), 878-880.

  • Vabulas, R. M., Ahmad-Nejad, P., da Costa, C., Miethke, T., Kirschning, C. J., Hacker, H., & Wagner, H. (2001). Endocytosed HSP60s use TLR2 and TLR4 to activate the TIR signaling pathway in innate immune cells. The Journal of biological chemistry, 276(33), 31332-31339.

  • Valdespino, V. M., & Valdespino, V. E. (2006). Cervical cancer screening: state of the art. Current opinion in obstetrics & gynecology, 18(1), 35-40.

  • Villa, L. L. (2008). Assessment of new technologies for cervical cancer screening. The Lancet Oncology.

  • Vizcaíno, J. A., Csordas, A., Del-Toro, N., Dianes, J. A., Griss, J., Lavidas, I., . . . Hermjakob, H. (2016). 2016 update of the PRIDE database and its related tools. Nucleic Acids Research, 44(D1), D447-D456.

  • Waggoner, S. E. (2003). Cervical cancer. In Lancet (Vol 361, bll 2217-2225).

  • Waldmann, T. A. (2003). Immunotherapy: Past, present and future. Nature Medicine.

  • Walsh, M. C., Kim, G. K., Maurizio, P. L., Molnar, E. E., & Choi, Y. (2008). TRAF6 autoubiquitination-independent activation of the NF-kB and MAPK pathways in response to IL-1 and RANKL. PLoS ONE, 3(12).

  • Waterston, R. H., Lindblad-Toh, K., Birney, E., Rogers, J., Abril, J. F., Agarwal, P., . . . Mouse Genome Sequencing, C. (2002). Initial sequencing and comparative analysis of the mouse genome. Nature, 420(6915), 520-562.

  • Wu, Y., Fan, Y., Xue, B., Luo, L., Shen, J., Zhang, S., . . . Yin, Z. (2006). Human glutathione S-transferase P1-1 interacts with TRAF2 and regulates TRAF2-ASK1 signals. Oncogene, 25, 5787-5800.

  • Wulfkuhle, J. D., Liotta, L. A., & Petricoin, E. F. (2003). Early detection: Proteomic applications for the early detection of cancer. Nat Rev. Cancer, 3(4), 267-275.

  • Ye, Z., Song, H., Higgins, J. P. T., Pharoah, P., & Danesh, J. (2006). Five glutathione S-transferase gene variants in 23,452 cases of lung cancer and 30,397 controls: Meta-analysis of 130 studies. PLoS Medicine, 3(4), 524-534.

  • Yokota, J. (2000). Tumor progression and metastasis. Carcinogenesis, 21(3), 497-503.

  • Zagouri, F., Sergentanis, T. N., Chrysikos, D., Filipits, M., & Bartsch, R. (2012). Molecularly targeted therapies in cervical cancer. a systematic review. Gynecologic Oncology.

  • Zuris, J. A., Thompson, D. B., Shu, Y., Guilinger, J. P., Bessen, J. L., Hu, J. H., Liu, D. R. (2014). Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo. Nature biotechnology, 33(1), 73-80.


Claims
  • 1. Antisense oligonucleotides to inhibit the expression of glutathione S transferase proteins, for the manufacture of a drug adapted for the treatment of cancer, to be administrable in mammals previously diagnosed with cancer.
  • 2. The antisense oligonucleotides according to claim 1, wherein the glutathione S transferases are GSTM3 and GSTP1.
  • 3. The antisense oligonucleotides according to claim 1, wherein GSTM3 and GSTP1 are used as therapeutic targets and/or prognostic factors.
  • 4. The antisense oligonucleotides according to claim 1, wherein said oligonucleotides include 15-50 nucleotides in length and bases 1-773 of GSTP1 and bases 1-4144 of GSTM3, with a similarity of 100-50% of both sequences.
  • 5. The antisense oligonucleotides according to claim 4, wherein said oligonucleotides are 18-30 nucleotides in length.
  • 6. The antisense oligonucleotides according to claim 4, wherein said oligonucleotides are 20-25 nucleotides in length.
  • 7. The antisense oligonucleotides according to claims 4, 5 and 6, wherein the bases for GSTP1 are close to the start codon and for GSTM3 close to the start codon.
  • 8. The antisense oligonucleotides according to claims 4, 5 and 6, wherein the similarity of both sequences is 100-80%.
  • 9. The antisense oligonucleotides according to claims 4, 5 and 6, wherein the similarity of both sequences is 100-90%.
  • 10. The antisense oligonucleotides according to claim 1, wherein the oligonucleotide is one having sugar modified bases, column or skeleton modifications, nucleobase modifications and general modifications in natural oligonucleotides.
  • 11. The antisense oligonucleotides according to claim 1, wherein the oligonucleotides are selected from the following:
  • 12. The antisense oligonucleotides according to claim 11, wherein the preferred oligonucleotides have the following sequences:
  • 13. The antisense oligonucleotides according to claim 1, wherein said oligonucleotides are directed to the messenger ribonucleic acids (mRNA) of the GSTM3 and GSTP1.
  • 14. Antisense oligonucleotides according to any one of the preceding claims, wherein at least one or more oligonucleotides combined, are used to specifically block one protein or both proteins.
  • 15. The antisense oligonucleotides according to claim 1, wherein the cancer is any wherein the tumor tissue contains one or both GSTM3 and GSTP1 proteins.
  • 16. The antisense oligonucleotides according to claim 1, wherein the cancer is selected for lung cancer, breast cancer, colorectal cancer, prostate cancer, stomach cancer, liver cancer, esophageal cancer, cervical cancer, thyroid cancer, bladder cancer, non-Hodgkin lymphoma, pancreatic cancer, leukemia, kidney cancer, uterine body cancer, oropharyngeal cancer, brain and central nervous system cancer, ovarian cancer, melanoma cancer, gallbladder cancer, laryngeal cancer, multiple myeloma cancer, nasopharyngeal cancer, laryngopharyngeal cancer, Hodgkin lymphoma, testicular cancer, salivary gland cancer, vulvar cancer, Kaposi sarcoma cancer, penile cancer, mesothelioma, and vaginal cancer.
  • 17. A kit for use in the identification of a subject to be treated with the oligonucleotides of claim 1, comprising at least one antisense oligonucleotide of glutathione S transferases, a protein extraction solution, at least two antibodies to the identification of proteins and optionally a secondary antibody, and a colorimetric developing solution for immunodetection assays such as: western staining, lateral flow membranes, ELISA or immunohistochemistry.
  • 18. The kit according to claim 17, wherein the proteins to be identified are the GSTM3 and GSTP1 proteins.
  • 19. A method for the identification of GSTs in vitro in samples of patients previously diagnosed with cancer comprising: a) extracting the protein from the tumor tissue, b) carrying out an analysis by immunodetection techniques and c) inhibiting the expression of the protein by use of the antisense oligonucleotide of claim 1.
  • 20. A method for the treatment of cancer comprising a) the identification of GSTs in vitro in samples of patients previously diagnosed with cancer, b) the extraction of the protein from the tumor tissue, c) carrying out an analysis by immunodetection techniques and d) administering an antisense oligonucleotide directed to the messenger ribonucleic acids (mRNA) of the GSTM3 and GSTP1.
Priority Claims (1)
Number Date Country Kind
MX/A/2019/001389 Jan 2019 MX national
PCT Information
Filing Document Filing Date Country Kind
PCT/MX2020/000003 1/30/2020 WO 00