WO2013126872A1 - Stratégie thérapeutique anti-cancéreuse pour combattre la résistance aux traitements contre le cancer et pour permettre la personnalisation de traitement de patients - Google Patents
Stratégie thérapeutique anti-cancéreuse pour combattre la résistance aux traitements contre le cancer et pour permettre la personnalisation de traitement de patients Download PDFInfo
- Publication number
- WO2013126872A1 WO2013126872A1 PCT/US2013/027600 US2013027600W WO2013126872A1 WO 2013126872 A1 WO2013126872 A1 WO 2013126872A1 US 2013027600 W US2013027600 W US 2013027600W WO 2013126872 A1 WO2013126872 A1 WO 2013126872A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- dnase
- gene
- promoter
- cells
- inhibitor
- Prior art date
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
- C07K14/4701—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
- C07K14/4747—Apoptosis related proteins
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
- A61K48/005—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N9/00—Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
- C12N9/14—Hydrolases (3)
- C12N9/16—Hydrolases (3) acting on ester bonds (3.1)
- C12N9/22—Ribonucleases RNAses, DNAses
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q1/00—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
- C12Q1/68—Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
- C12Q1/6876—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
- C12Q1/6883—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
- C12Q1/6886—Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Y—ENZYMES
- C12Y301/00—Hydrolases acting on ester bonds (3.1)
- C12Y301/21—Endodeoxyribonucleases producing 5'-phosphomonoesters (3.1.21)
- C12Y301/21001—Deoxyribonuclease I (3.1.21.1)
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
- A61K48/005—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
- A61K48/0058—Nucleic acids adapted for tissue specific expression, e.g. having tissue specific promoters as part of a contruct
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/01—Fusion polypeptide containing a localisation/targetting motif
- C07K2319/09—Fusion polypeptide containing a localisation/targetting motif containing a nuclear localisation signal
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2830/00—Vector systems having a special element relevant for transcription
- C12N2830/008—Vector systems having a special element relevant for transcription cell type or tissue specific enhancer/promoter combination
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Q—MEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
- C12Q2600/00—Oligonucleotides characterized by their use
- C12Q2600/112—Disease subtyping, staging or classification
Definitions
- the present invention relates to therapeutics and methods of treating cancer.
- the present invention relates to modified programmed-cell-death executioner genes.
- DNA-degrading enzymes are important mediators of aptoptosis.
- DNase 1 for example, is a powerful DNA-degrading enzyme, that preferentially cleaves DNA at phosphodiester linkages adjacent to a pyrimidine nucleotide, producing tetranucleotides, and also binds actin.
- DNase 1 acts on both single- and double-stranded DNA as well as chromatin.
- DNase 1 is the major extracellular DNase protein and a major executioner of intracellular apoptosis, i.e.
- DNase 1 could have access to the cell's DNA only after the activated apoptosis cascade led to initial disintegration of the nucleus membrane.
- Various combinations of these four mechanisms likely protect normal and cancerous cells from additional DNA-degrading enzymes to DNAse I.
- Napirei, et al. deleted the signal peptide from DNase 1 and observed localization of DNase 1 in both the cytoplasm and nucleus, but no apoptosis.
- Linardou, et al. created a scFv-DNase 1 chimera against human placental alkaline phosphatase (hPLAP) that decreased cell viability when measuring the level of DNA synthesis in a tritium-labeled thymidine (3H-TdR) incorporation assay.
- Bovine DNase 1 was used. The signal peptide was not removed, and an NLS was not included. The actin binding site was not mutated, and therefore, the chimera was not resistant to DNase 1 inhibition by actin. Apoptosis tests were not performed, only cytotoxicity was tested. The scFv-Ab seemed to be cytotoxic as well, suggesting necrosis in addition to possible apoptosis.
- the actin binding site on DNase 1 has been inactivated to increase the potency of DNase 1 to dilute viscosity of upper respiratory secretions in Cystic Fibrosis (CF) patients (Ulmer, et al., PNAS 1996, 93:8225-9; Pan, et al., J Biol Chem 1998, 273:18374-81 ) and for treatment of systemic lupus erythematosus (Pan, et al., J Biol Chem 1998, 273:18374-81 ).
- Recombinant human DNase 1 (PULMOZYME®, Genentech) was approved by the FDA in December 1998 as an aerosolized mucolytic agent for the management of patients with CF.
- DNase 1 is safe to administer to humans. Ulmer, et al. showed that the binding site for actin was mutated and increased DNAse 1 's ability to digest DNA in sputum, thus, reducing its viscosity. The DNase 1 was not aimed at functioning inside cells or inducing apoptosis in cells. The signal peptide, which diverts the protein away from the nucleus, was not removed, and an NLS was not included.
- Malignant melanoma is the most aggressive form of cancer. Its dimensions are reported in millimeters. Tumor thickness approaching 4 mm presents a high risk of metastasis. A diagnosis of metastatic melanoma carries with it a median survival of 6-9 months. At the time of diagnosis, 20% of patients have metastasis; 16% to the lymph nodes and 4% to the distal organs. Current methods of treatment include surgical resection, radiation, chemotherapy with dacarbazine, tamoxifen, or temozolamide, or immunotherapeutics such as IL-2, IFN-a. Only a minority of patients respond to these methods of treatment.
- Some of the therapeutics that have been developed as specific molecularly targeted therapies include immunotherapies such as vaccines (e.g. Hi-8TM MEL-vaccine (Oxxon Therapeutics)) and ONCOVEX® (BioVex, Inc.), as well as biotherapeutics that perform functions such as targeting anti-apoptosis/resistance (e.g. Bcl2, IAP, NF-KB inhibition), promoting apoptosis (e.g. TRAIL ligands), siRNA (e.g. against Mitf), and suicide gene-therapy (e.g. HSV-tk).
- immunotherapies such as vaccines (e.g. Hi-8TM MEL-vaccine (Oxxon Therapeutics)) and ONCOVEX® (BioVex, Inc.), as well as biotherapeutics that perform functions such as targeting anti-apoptosis/resistance (e.g. Bcl2, IAP, NF-KB inhibition), promoting apoptosis (e.g. TRAIL
- Gene therapy i.e. the transfer of genetic material into living cells for treatment or prevention of disease, has grown in popularity for use as a treatment over the years.
- the United States conducts the most gene therapy clinical trials, with some conducted in Europe and Asia.
- the United States also leads the rest of the world in the study of malignant melanoma with gene therapy clinical trials.
- Gene therapy targeted to melanoma cells involves the introduction of "suicide” genes, such as a herpes simplex virus thymidine kinase gene (HSVtk), the transfer of tumor suppressor genes such as p53 gene or p16INK4a, the inactivation of oncogenic signaling pathways such as ras, c-myc, and signal transducers and activators of transcription-3 (Stat3), and the introduction of genes encoding immunologically relevant molecules such as allogenic MHC class I genes, cytokine genes (GM-CSF, IL-2, IFNs), and co-stimulatory molecules (B7.1 ). Gene therapy can also be targeted to the host's immune cells.
- suicide genes such as a herpes simplex virus thymidine kinase gene (HSVtk)
- tumor suppressor genes such as p53 gene or p16INK4a
- Stat3 signal transducers and activators of transcription-3
- Gene therapy can also be targeted to
- T cells are targeted with the introduction of neomycin phosphotransferase gene and chimeric receptor (IL-2R/GM- CSFR).
- Dendritic cells are targeted with genes encoding melanoma antigens (MART- 1 /Melan A) and CD40 ligand.
- GLYBERA® (alipogene tiparvovec, Amsterdam Molecular Therapeutics) is an AAV-1 vector encoding lipoprotein lipase for treatment of patients with lipoprotein lipase deficiency.
- INGN- 241 (Introgen) is an E1 -deleted, replication-incompetent adenoviral vector encoding melanoma-differentiation-associated gene-7 (mda-7; interleukin-24) for treatment of metastatic melanoma.
- TNFerade is an E1 -, E3-, and E4-delected andenoviral vector encoding human TNF-a under the control of the radiation-inducible early growth response promoter for use in pancreatic cancer.
- a retrovirus by MolMed encoding herpes simplex virus thymidine kinase transduced ex vivo into hematopoietic stem cells is used for graft-versus-host disease.
- ALLOVECTIN-7® velimogene aliplasmid, Vical is a DNA plasmid encoding the human leukocyte antigen-B7 (HLA-B7) and 2-microglobulin complex in the context of cationic lipid mixture (DMRIE/DOPE) for use in chemotherapy-na ' ive patients with metastatic melanoma.
- PROSAVIN® (Oxford Biomedica) is combined lentivirus and equine infectious anemia virus vectors encoding aromatic amino acid decarboxylase, tyrosine hyroxylase and GTP-cyclohydrolase-1 used for Parkinson's disease.
- tgAAC-94 (Targeted Genetics) is an AAV-2 encoding lgG1 Fc and the TNF-a receptor used in rheumatoid arthritis.
- Anti-TRAILFM agonistic antibody Human Genome Sciences/Cambridge Antibody Technology/Takeda
- Anti-TRAILR2 agonistic antibody Human Genome Sciences/Cambridge Antibody Technology
- TRAIL Gener/Amgen
- GENASENSE® oblimersen, antisense oligonucleotide targeting BCL2, Genta, Inc. was used in malignant melanoma, multiple myeloma, and chronic lymphocytic leukemia (CLL).
- SPC-2996 antisense oligonuleotide targeting BCL2, Santaris Pharma
- AT 101 ((-)-Gossypol, Ascenta Therapeutics Inc.)
- small molecule BCL2-family inhibitor Gamin X Biotech
- apoptosis- inducing agents include ABT-737 (small molecule BCL2-family inhibitor, Abbott Laboratories/ldun Pharmaceuticals), IPI-983L/IPI-194 (small molecule BCL2-family inhibitors, Infinity Pharmaceuticals), XIAP-BIR2 inhibitor (Burnham Institute), XIAP-BIR3 inhibitor (UT Southwestern), XIAP-BIR3 inhibitor (Abbott Laboratories), and Nutlins (MDM2 inhibitors, Wyeth).
- agents potentiate the mitochondrial pathway and decrease BCL2, BCL-XL, and MCL-1 (Antisense to BCL2, BCL-XL, and MCL-1 ), downregulate BCL2, BCL-XL, and MCL-1 (MEK1 inhibitors, PD98059), damage mitochondria (cisplatin/doxorubicin), and increase apoptosis activating factor (APAF) levels by inhibiting methylation (5-aza-deoxycytidine).
- Agents inhibit NF- ⁇ activation by proteasome inhibition and upregulation of ⁇ (PS341 ), and competing with p53 for co- transcriptional factors (temozolomide and vinblastine).
- agents downregulate inhibitor of apoptosis protein (IAP) levels by decreasing XIAP levels (actinomycin-D, fludarabine, second mitochondria-derived activator of caspase (SMAC)/DIABLO constructs).
- Apoptosis is a physiological mechanism of programmed cell death by which unwanted cells are eliminated from tissues in response to specific stimuli.
- the apoptosis cascade is shown in FIGURE 1 .
- the current failure of the above described gene therapy treatments results from the fact that the majority of cancers protect themselves by inactivating or underexpressing death receptors, not expressing or inactivating intermediate components of the apoptosis cascade, and overexpressing anti-apoptotic proteins that inactivate pro- apoptotic components along the apoptosis cascade as shown in FIGURE 2. Therefore, a method of treatment is needed that overcomes the difficulties presented by the natural protection mechanisms of cancer cells against the apoptosis cascade.
- the present invention provides for a gene construct, including a programmed- cell-death executioner gene having a nuclear localization signal, a deleted signal peptide, an inhibitor-resistant binding site, a promoter, and an activator.
- the gene construct is a late player in the cell death process that has the ability to initiate programmed cell death and execute it with increased efficiency.
- the present invention provides for a method of making the gene construct by modifying a nuclease encoding gene, which in its native form and under physiological conditions is incapable of triggering cell death by adding a nuclear localization signal, deleting a signal peptide, mutating a binding site for an inhibitor to make it resistant to that inhibitor, adding a promoter for exclusive expression in selected cells, and adding an activator.
- the present invention also provides for a method of eliminating undesired cells from a patient, including the steps of administering a therapeutic containing a gene construct, delivering the therapeutic to undesired cells, activating apoptosis of the undesired cells without triggering the apoptosis cascade, and destroying the undesired cells.
- the present invention provides for a method of treating cancer, including the step of activating apoptosis of cancer cells without the need for triggering the apoptosis cascade.
- the present invention provides for an array that is generated of at least two gene constructs described above, with all of the constructs differing with respect to the programmed-cell-death executioner gene and with respect to the nuclear localization signal.
- the present invention also provides for a method of personalizing anti-cancer treatment by taking a sample of cancerous cells from a patient, applying the cells to an array of at least two gene constructs with all of the constructs differing with respect to the programmed-cell-death executioner gene, with respect to the promoter and with respect to the nuclear localization signal, selecting the gene construct with the highest sensitivity to the cancerous cells, and treating the patient with the selected gene construct.
- the present invention further provides for using the array as a diagnostic device to determine if a patient has cancer by taking a sample of cells in a patient, determining if a gene construct in the array is sensitive to the cells, and if cells are sensitive to at least one gene construct, determining that the patient has cancer.
- the present invention also provides for a method of increasing DNase 1 resistance to actin binding by mutating native amino acids at an actin binding site chosen from the group consisting of Glu-13, His-44, Asp-53, Tyr-65, Val-66, Val-67, Glu-69, Ala-1 14, and combinations thereof.
- the present invention provides for a method of increasing catalytic activity of DNase 1 by mutating native amino acids to change the electrical charge chosen from the group consisting of Gln-9, Glu-13, Asn-74, and combinations thereof.
- FIGURE 1 is a diagram of the apoptosis cascade
- FIGURE 2 is a diagram of how cancer cells protect themselves from the apoptosis cascade
- FIGURE 3 is a diagram of how DNase 1 is prevented from activating in a cell
- FIGURE 4 is a diagram of the building blocks of a gene construct of the present invention.
- FIGURES 5A and 5B are schematic representations of the designed gene constructs, wherein 5A shows the genetic alterations used for generating the DNase 1 constructs, and 5B shows the composition of the expressed DNase 1 protein constructs (Cytomegalus virus promoter (CMV), Signal peptide (SP), Deoxyribonuclease-1 gene (DNase 1 ), nuclear localization signal (NLS), green fluorescent protein (GFP), mutation of an actin binding site (X on left side of DNase 1 symbol), mutation of a proteolytic activity site (X on right side of DNase 1 symbol)); [00035] FIGURES 6A and 6B are assessments of DNase 1 gene construct expression at the molecular level by Quantitative Real Time Reverse Transcription PCR (QRT-PCR) and Western Blotting with mRNA (6A) and protein (6B) (SP - Signal peptide; NLS - Nuclear Localization Signal; Cat-G - Cat- GFP; GFP - Green fluorescence protein; WT - Wild
- FIGURE 7 is a graph of in vitro assessment of DNase 1 activity by a colorimetric assay, values are presented relative to Wild-type DNase 1 , open rectangles represent means and lines represent 95% confidence intervals. WT - Wild type DNase 1 ;
- FIGURES 8A-8D are photographs of representative samples of localization study of DNase 1 protein constructs in Mel-Juso human melanoma cells 24 hours after transfection
- FIGURE 8A shows that addition of a NLS to the GFP tagged DNase 1 (C01 ) did not change the characteristic cytoplasmic distribution of the secretory protein
- FIGURE 8B shows elimination of the Signal peptide enabled targeting the NLS-fused DNase 1 to the nucleus
- FIGURES 8C and 8D are controls, closed arrow heads point to nuclear distribution, open arrow heads point to scarce or absent cytoplasmic distribution (SP - Signal peptide, NLS - Nuclear Localization Signal, GFP - Green fluorescence protein; Original magnification x 400);
- FIGURES 10A-10D are fluorescence and phase contrast microscopy photographs of Mel-Juso human melanoma cells using TUNEL assay; DNase 1 gene constructs C1 1 (10C) and C13 (10D) as well as controls (10A and 10B), arrows indicate apoptotic cells; Original magnification x 400;
- FIGURE 1 1 is a photograph of a DNA fragmentation assay (SP - Signal peptide; NLS - Nuclear Localization Signal; AS - Activity site; Mark. - 200 bp Marker; Untreat. - Untreated cells; WT - Wild type DNase 1 );
- FIGURE 12A shows a representative LSC-generated scattergraph with increased proportion of cell population undergoing apoptosis (high green fluorescence intensity) after treatment with NLS+Actin-resistant DNase 1 (C1 1 ), as compared to treatment with activity knocked down DNase 1 (C18), Wild-type DNase 1 or Mock
- FIGURE 12B is a graph showing percent of TUNEL positive (apoptotic) cells quantified by LSC, wherein means are represented by bars and numerical values, the S.E.M. for C1 1 , C13, Wild Type and C18 were 4.6%, 1 .2%, 0.8% and 0.6%, respectively, and for all other samples ⁇ 0.3%.
- FIGURE 13 is an index of DNase 1 gene constructs design and generation
- FIGURE 14 is a list of identification of the DNase 1 gene constructs
- FIGURE 15 is a flow chart of DNase 1 gene constructs design
- FIGURE 16 is a flow chart of DNase 1 gene constructs design
- FIGURE 17 is a flow chart of DNase 1 gene construct mutagenesis primer usage
- FIGURE 18 is a flow chart of DNase 1 gene construct mutagenesis primer usage
- FIGURE 19 is a flow chart of DN1 transfer from pDONR223 to pDEST-47;
- FIGURE 20 is a flow chart of mutagenesis sites;
- FIGURE 21 is a flow chart of mutagenesis sites
- FIGURE 22 is a flow chart of mutagenesis sites
- FIGURE 23 is a flow chart of mutagenesis sites
- FIGURE 24 is a flow chart of mutagenesis sites
- FIGURE 25 is a flow chart of mutagenesis sites
- FIGURE 26 is a flow chart of mutagenesis sites
- FIGURE 27 is a flow chart of mutagenesis sites
- FIGURE 28 is a flow chart of mutagenesis sites
- FIGURE 29 is a flow chart of mutagenesis sites
- FIGURE 30 is a flow chart of mutagenesis sites
- FIGURE 31 is a flow chart showing from C.14 generation of C.01 ;
- FIGURE 32 is a table showing sequences of primers.
- FIGURE 33 is a table showing sequences of primers.
- the present invention provides a treatment for cancer and various chronic and infectious diseases, based on modified programmed-cell-death executioner genes.
- a programmed-cell-death executioner protein is a DNA-degrading enzyme.
- the modified programmed-cell-death executioner gene is a core enzyme that lacks a nuclear localization signal and has the ability to degrade DNA.
- a modified DNA- degrading enzyme most preferably a modified human recombinant (hr) deoxyribonuclease-1 (hr DNase 1 ) gene is used.
- modified programmed-cell-death executioner genes can be used, such as, but not limited to, DNase 1 L1 , DNase 1 L2 (DHP1 ), DNase 1 L3 (DNase ⁇ ; DHP2), DNase 2A, DNase 2B, Caspase-activated DNase (CAD), Endonuclease G (ENDOG), Granzyme B (GZMB), and DNase X.
- DNase 1 L1 DNase 1 L2
- DNase 1 L3 DNase 1 L3
- DNase 2A DNase 1 L3
- DNase 2B DNase 2A
- DNase 2B Caspase-activated DNase
- CAD Endonuclease G
- GZMB Granzyme B
- DNase X DNase X
- Gene construct refers to the programmed-cell-death executioner gene as modified by the method of the present invention, and each term can be used interchangeably herein.
- the gene constructs of the present invention are made by performing the following modifications on a programmed-cell-death executioner gene such as a gene for a DNA-degrading enzyme, generally shown in FIGURE 4 for the specific example of DNase 1 .
- a programmed-cell-death executioner gene such as a gene for a DNA-degrading enzyme, generally shown in FIGURE 4 for the specific example of DNase 1 .
- Each of these modifications serve to prevent premature activation of cell death by the programmed-cell-death executioner gene by several mechanisms. These mechanisms include the barrier function of the nuclear membrane, the presence of a signal peptide that diverts the programmed-cell-death executioner gene from the nucleus, the absence of a nuclear localization signal, and the presence of an inhibitor, actin, in the nucleus, endoplasmic reticulum, and cytoplasm, as shown in FIGURE 3.
- the programmed-cell-death executioner gene is first modified by equipping it with a (5'-/-3') nuclear localization signal (NLS) to render access of the gene to the nucleus.
- NLS nuclear localization signal
- PPKKKRKV SEQ ID NO: 1
- KRPAATKKAGQAKKKKL SEQ ID NO: 2
- NLS-1 KR in FIGURE 27 and primers in FIGURE 33 is employed in C27-26p.
- NAPRGKKPAPG (SEQ ID NO: 3) (see NLS-2NA in FIGURE 27 and primers in FIGURE 33) is employed in C28-26p.
- RKFKKKFNK (SEQ ID NO: 4) (see NLS-3RK in FIGURE 27 and primers in FIGURE 33) is employed in C29-26p.
- SRKRPRP (SEQ ID NO: 5) (see NLS-4SR in FIGURE 27 and primers in FIGURE 33) is employed in C30-26p. Any other suitable NLS can be used and can be added by one skilled in the art.
- the signal peptide is deleted to prevent loss of the gene construct by diversion to the endoplasmic reticulum. This increases killing efficiency of the gene construct.
- the binding site for actin (the inhibitor) is mutated or deleted to make the gene construct actin-resistant, thus enabling the gene construct to digest a cell's DNA. This eliminates inhibition.
- a promoter is added and secures exclusive expression of the killer gene in selected cell types, such as malignant melanoma cells.
- the cell types can be cancerous or otherwise undesired cells.
- the method of the addition is explained further below in the Example (Materials and Methods). This provides selective targeted expression.
- the promoter can include a melanocyte-specific promoter such as tyrosinase, melanoma inhibitory activity (MIA), SILV/PMEL17/GP100, Melan-A/MART-1 , melanocortin-1 receptor (MC1 R) and microphthalmia-associated transcription factor (MITF).
- the promoter can be from prostate-specific antigen (PSA), prostate-specific membrane antigen (PSMA), prostatic acid phosphatase (PAP), ARR2PB, PSA enhancer - rat probasin promoter, 1455 bp PSA enhancer upstream to PSA or hKLK2, or prostate-specific chimeric enhancer PSES.
- PSA prostate-specific antigen
- PSMA prostate-specific membrane antigen
- PAP prostatic acid phosphatase
- ARR2PB PSA enhancer - rat probasin promoter
- 1455 bp PSA enhancer upstream to PSA or hKLK2 or prostate-specific chimeric enhancer PSES.
- the promoter can be hSCGB2A2 (for mammary carcinoma cells), DF3/MUC1 , DF3 enhancer, or DF3 enhancer - DF3 promoter.
- the promoter can be from thyroglobulin or calcitonin (CT).
- the promoter can be a combination of the TTF1 gene driven by the hTERT promoter with 5 tandem copies of a portion of hSPA1 , or hexokinase II.
- the promoter can be liver-selective phosphoenolpyrovate carboxykinase, or albumin gene promoter.
- the promoter can be RIP (rat insulin promoter) activated only in ⁇ cells.
- the promoter can be (1 ) targeting glioma: GFAP, Musashi, gfa 2, NSE, and (2) targeting glioma or glioblastoma: placing Nestin's 2nd intron before the 5' upstream region (2iNP).
- the promoter can be HLA-Dralpha, CD4, CD19, or Ig kappa.
- the promoter can be the HIV-1 long terminal repeat (LTR) promoter. Any other suitable promoter can be used.
- An on/off switch i.e. activator
- the activators can include a promoter (in addition to the promoter above used for selective expression).
- the gene construct can be packaged in an envelope made of nanoparticles or from viruses and which has on its surface receptors that direct the envelope and its content to the selected cells.
- an envelope made of nanoparticles or from viruses and which has on its surface receptors that direct the envelope and its content to the selected cells.
- Nanoparticle or viral envelopes are delivery mechanisms used to encapsulate therapeutic treatments in order to cause the treatment to pass through tumorous tissues, but not pass through healthy tissue.
- the outside of the envelope is generally covered with molecules that are recognized by receptors present on tumor cells or infectious cells but not on healthy cells. This way, the therapeutic can more specifically be delivered to a tumor to achieve therapeutic concentrations of the gene constructs at the target tissue and to reduce potential damage to healthy tissue.
- the envelope includes surface receptors that are specific for melanocytic and melanoma cells in order to treat melanoma.
- the envelope can be designed to be targeted to any cells desired to be eliminated.
- the gene constructs of the present invention can be delivered in the form of a naked DNA plasmid or in various microbial vectors known in the art, including, but not limited to, retrovirus vectors, adeno-associated virus vectors, lentivirus vectors (Walther, et al., Drugs 2000 60:249-71 ), adenovirus vectors, vaccinia virus vectors, poxvirus vectors, and virus-like particles (Harrop, et al., 2006). Attenuated bacterial vectors can also be employed, such as species of Salmonella, Shingella, Listeria, Yersinia, and Escherichia (Vassaux, et al., J Pathol.
- Cells can also be used as therapeutic carriers, including, but not limited to lymphocytes, neutrophils, monocytes, and stem cells (Roth, et al., Gene Ther. 2008 May;15(10):716-29).
- One very important advantage of the present invention is the ability of the gene constructs to activate apoptosis without triggering the apoptosis cascade that would normally be triggered in classical apoptosis-inducing therapies such as suicide gene therapy.
- the gene constructs of the present invention bypass all negative and positive inhibitory feedbacks that generally exist with programmed-cell-death executioner genes in order to destroy a targeted cell.
- the negative and positive inhibitory feedbacks are present at almost every level along the apoptosis cascade.
- the gene constructs of the present invention deliver the final effector of apoptosis, activated DNases, to their normal site of action, the nucleoplasm.
- One benefit of using the gene constructs of the present invention is that the number of patients that are able to respond to anti-cancer treatment is increased because the gene constructs of the present invention bypass many of the main treatment-resistant mechanisms of cancer, i.e. the apoptosis cascade. This is accomplished by triggering the chain of morphological, cancer-cell disintegration events from the last component of the apoptosis cascade.
- suicide gene therapies for cancer that use activating death-receptors (CD95, tumor necrosis factor (TNF)) at the beginning of the apoptosis cascade, or overexpressing intermediate apoptosis-cascade/cell-cycle components (Caspases, p53), which are dependent on an intact down-stream chain of components.
- the gene constructs of the present invention can therefore be used in patients that are resistant to apoptosis-inducing gene therapies such as suicide gene therapy.
- the gene constructs of the present invention are able to be used in patients that are resistant to other apoptosis-inducing treatments such as radiotherapy and chemotherapy. Therefore, a method of treating cancer (or any other disease as described herein) is provided by activating apoptosis of cancer cells (or any other desired cells) without triggering the apoptosis cascade.
- the gene constructs can be used to target any disease that has specific markers in the diseased cells.
- the gene construct can be targeted to promoters that are present in diseased cells, so that they can be selectively eliminated, leaving healthy cells intact.
- the gene constructs can be used to treat many different kinds of cancers, preferably malignant melanoma, but also prostate cancer, lymphoma, leukemia, throat, pancreatic, thyroid, ovarian, neuroendocrine (small cell lung cancer, neuroblastoma, carcinoid), or medullary thyroid cancer.
- the cancer is in a tissue or organ that possesses a specific promoter/enhancer that can be used to target the gene construct's expression.
- melanocytic cells In malignant melanoma, only melanocytic cells contain promoters for genes involved in the synthesis of the melanin pigment.
- the gene constructs can also be used to treat infectious diseases such as, but not limited to, HIV, AIDS, malaria, and Leishmania.
- the gene construct can also be used to eliminate specific cells, such as, but not limited to, subtypes of immune cells that are involved in the etiology of chronic inflammatory diseases such as rheumatoid arthritis, inflammatory bowel disease (Crohn's disease, ulcerative colitis), or graft- versus-host disease (GVHD). Any disease that involves specific types of T cells can also be targeted. While cancer cells are specifically discussed, it should be understood that the methods herein apply to any cells desired to be eliminated from a patient, provided that they produce a unique protein to the targeted tissue/organ.
- a method of eliminating undesired cells from a patient including the steps of administering a therapeutic containing a gene construct, delivering the therapeutic to undesired cells, and destroying the cells.
- this method is used to treat cancer, especially malignant melanoma, and the undesired cells are cancerous or tumorous cells.
- the gene construct is administered in the envelope as described above, through the use of a gene gun or injection. Other methods of administration are further described below.
- the gene construct is preferentially delivered to the undesired cells and not healthy cells due to the surface receptors that direct the envelope and its content to the undesired cells.
- the gene constructs are able to gain access to the nucleus due to their modifications as described above and destroy the cell.
- removing the signal peptides prevents the cell from packing the gene construct in an endosome and shipping it out of the cell (secreting the gene construct).
- Adding an NLS provides the gene construct an access into the nucleus; hence, it can come in vicinity to the target DNA in order to defragment it. Knocking down the binding site for actin prevents actin from inhibiting the gene construct.
- a method is provided of treating malignant melanoma by administering a therapeutic containing a genetically modified DNA-degrading enzyme at the core of a gene construct, such as a hrDNase 1 gene construct, delivering the therapeutic to melanoma cancer cells, and destroying the melanoma cancer cells.
- the treatment is advantageous over other prior art treatments, because the gene constructs include components that limit the expression to targeted cells and not non-targeted cells, and the envelope used for delivery of the gene construct includes surface homing receptors that target the gene construct to the tissue of choice, thereby allowing the gene construct to accumulate in the targeted tissue and further reducing side effects for the patient, as further described below.
- the therapeutics of the present invention enable treatment of cancer in patients with a compromised immune system, or even a severely compromised immune system, since they do not require the participation of the immune system. This is in contrast to current cancer therapies that are immunotherapeutic approaches and dependent on the recruitment of a patient's immune system in order to fully function. A substantial number of patients having cancer metastases also have deficiencies in their immune system functions. The present invention can therefore treat more patients than other current therapies due to the fact that a fully functioning immune system is not required for effectiveness of the therapeutics herein.
- a method is provided of treating a cancer patient having a compromised immune system, by administering a therapeutic containing a gene construct to a patient having a compromised immune system, delivering the therapeutic to tumor cells, destroying the tumor cells, and treating cancer.
- Another advantage to the present invention is that it has fewer side effects than current cancer therapies, since the present invention enables the elimination of cancer cells without a significant damage to neighboring healthy cells.
- treatment with the modified programmed-cell-death executioner genes of the present invention did not show any killing of non-transfected cells.
- suicide gene-therapy approaches such as HSV-tk are dependent on non-direct killing (bystander effect) due to low efficiency in direct killing.
- Suicide genes encode a protein that activates a cytotoxic prodrug. The activated drug can leak or be secreted to kill nearby nontransfected bystander cells.
- the lack of bystander effect in a treatment for cancer is important because the bystander effect of HSV-tk has been shown to be associated with damage to neighboring healthy cells and with death in treated animals.
- the present invention therefore provides a larger margin of safety for cancer patients, as healthy cells are protected.
- the gene constructs also avoid necrosis elicited by toxin- conjugated Abs.
- Necrosis is the premature death of cells that is caused by external factors, such as trauma, infections, or toxins. The content of dying cells reaches the intercellular space and causes a strong immune reaction that increases the damage to the tissue by killing healthy cells, which were not subject to the original insult.
- apoptosis is a physiological, intracellular mechanism that allows the elimination of dying cells without provoking harmful immunogenic reactions.
- a method is provided of treating cancer and reducing side effects, by administering a therapeutic containing the gene construct, preferentially delivering the therapeutic to tumor cells and not healthy cells, destroying the tumor cells, and treating cancer.
- An array is also provided in the present invention that is generated of at least two gene constructs each having different killer genes at their core, such as a gene encoding a DNA-degrading enzyme, different promoters and different nuclear localization signals.
- This enables personalized treatment of a patient having a tumor by treating this patient with a gene construct containing a promoter that can be active within a patient tumor and treating the patient with the gene-construct expressing the highest killing efficiency for the tumor.
- Cancer cells obtained from diagnostic biopsy on a patient, are grown in the laboratory and exposed to the various gene constructs to determine sensitivity level. The patient is then treated with the gene construct expressing the highest killing efficiency.
- the array can also include gene constructs that have different promoters as previously listed above. This array is useful in determining what type of cancer or disease a patient has.
- One example of an array is as follows. DNase 1 , DNase 1 -L2, DNase 2B, DNase X can be included and use any of the 5 NLSs mentioned above, in LD1 . Hence, there are 20 different gene constructs that can be prepared by these combinations, each of which can be included in an array.
- a method of personalizing anti-cancer treatment is provided by taking a sample of cancerous cells from a patient, applying the cells to an array of gene constructs in which all of the constructs differ with respect to the programmed-cell-death executioner gene and with respect to the nuclear localization signal, selecting the gene construct with the highest sensitivity to the cancerous cells, and treating the patient with the selected gene construct.
- the patient is treated as described above, enclosing the gene construct in the envelope, delivering the gene construct to cancerous cells, and destroying the cells.
- This method can also be practiced with any other cells that are desired to be eliminated from the patient, in order to select the best method of treatment.
- More than one gene construct can be used to treat the patient, depending on how many gene constructs in the array respond to the sample of the patient's cells. This is useful because many cancers evolve and change as they progress and thus targeting only one type of cell will not eradicate the cancer. Multiple gene constructs can therefore be used to fully treat the patient.
- the array can also be used as a diagnostic device to determine if a patient has cancer by taking a sample of cells in a patient, determining if a gene construct in the array is sensitive to the cells, and if cells are sensitive to at least one gene construct, determining that the patient has cancer.
- the array includes gene constructs with different promoters as well as different killer genes and NLSs. This can be a preliminary determination, and the diagnosis can prompt the patient to get further testing.
- the use of the array as a diagnostic device can also be used to diagnose a patient with other infectious diseases.
- the gene construct of the present invention can further include various mutations to modify properties of the gene construct.
- a preferred type of mutation is the mutation of an inhibitor-binding site of an enzyme such as a DNA-degrading enzyme, to render the enzyme less sensitive to its inhibitors.
- DNase 1 resistance to actin binding can be increased by mutating native amino acids at an actin binding site of the DNase 1 gene at Glu-13, His-44, Asp-53, Tyr-65, Val-66, Val-67, Glu-69, Ala-1 14, and combinations thereof.
- Other examples of actin binding sites that can be changed are listing in TABLE 1 below.
- the catalytic activity of DNA-degrading enzyme can be increased by mutation of appropriate amino acids.
- the catalytic activity of DNase 1 can be increased by mutating native amino acids of the DNase 1 gene to change the electrical charge of Gln-9, Glu-13, Asn-74, and combinations thereof.
- NLS Nuclear localization signal
- nucleus-directed construct including a DNA-degrading enzyme sensitive to a nuclear inhibitor other than actin will show increased killing efficiency when the relevant inhibitor-binding site is mutated to reduce inhibitor binding.
- the gene constructs of the present invention are administered and dosed in accordance with good medical practice, taking into account the clinical condition of the individual patient, the site and method of administration, scheduling of administration, patient age, sex, body weight and other factors known to medical practitioners.
- the pharmaceutically "effective amount" for purposes herein is thus determined by such considerations as are known in the art. The amount must be effective to achieve improvement including but not limited to improved survival rate or more rapid recovery, or improvement or elimination of symptoms and other indicators as are selected as appropriate measures by those skilled in the art.
- the compounds of the present invention can be administered in various ways. It should be noted that they can be administered as the compounds and can be administered alone or as an active ingredient in combination with pharmaceutically acceptable carriers, diluents, adjuvants and vehicles.
- the compounds can be administered orally, subcutaneously or parenterally including intravenous, intraarterial, intramuscular, intraperitoneal ⁇ , intratonsillar, and intranasal administration as well as intrathecal and infusion techniques. Implants of the compounds are also useful.
- the patient being treated is a warm-blooded animal and, in particular, mammals including man.
- the pharmaceutically acceptable carriers, diluents, adjuvants and vehicles as well as implant carriers generally refer to inert, non-toxic solid or liquid fillers, diluents or encapsulating material not reacting with the active ingredients of the invention.
- the doses can be single doses or multiple doses over a period of several days.
- the treatment generally has a length proportional to the length of the disease process and drug effectiveness and the patient species being treated.
- the pharmaceutical formulations suitable for injection include sterile aqueous solutions or dispersions and sterile powders for reconstitution into sterile injectable solutions or dispersions.
- the carrier can be a solvent or dispersing medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
- Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
- Nonaqueous vehicles such as cottonseed oil, sesame oil, olive oil, soybean oil, corn oil, sunflower oil, or peanut oil and esters, such as isopropyl myristate, may also be used as solvent systems for compound compositions.
- various additives which enhance the stability, sterility, and isotonicity of the compositions including antimicrobial preservatives, antioxidants, chelating agents, and buffers, can be added.
- antibacterial and antifungal agents for example, parabens, chlorobutanol, phenol, sorbic acid, and the like.
- isotonic agents for example, sugars, sodium chloride, and the like.
- Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin. According to the present invention, however, any vehicle, diluent, or additive used would have to be compatible with the compounds.
- Sterile injectable solutions can be prepared by incorporating the compounds utilized in practicing the present invention in the required amount of the appropriate solvent with various of the other ingredients, as desired.
- a pharmacological formulation of the present invention can be administered to the patient in an injectable formulation containing any compatible carrier, such as various vehicle, adjuvants, additives, and diluents; or the compounds utilized in the present invention can be administered parenterally to the patient in the form of slow-release subcutaneous implants or targeted delivery systems such as monoclonal antibodies, vectored delivery, iontophoretic, polymer matrices, liposomes, and microspheres. Examples of delivery systems useful in the present invention include those disclosed in U.S. Patent Nos.
- Dulbecco-S Phosphate-buffered saline PBS
- RPMI 1640 medium Fetal Calf Serum
- FCS Fetal Calf Serum
- L-Glutamine 200 mmol 0.5% Trypsin/5.3 mm EDTA were purchased from Fisher (Fairlawn, NJ), and Invitrogen (Grand Island, NY).
- Dimethyl sulfoxide DMSO from Sigma (St. Louis, MO).
- Cells were plated in 6 well plates at a density of 8 x 105 cells/well for TUNEL (Terminal transferase dUTP Nick-End Labeling) and DNA fragmentation assays, at a density of 4 x 105 cells/well for Western blot and at a density of 600 cells per well for colony forming assay (CFA). After 24 hours, the cells were transfected with 2 g of DNA (gene construct) using Lipofectamine LTX and Plus reagent (Invitrogen, Carlsbad, CA) according to the manufacturer's instructions. The transfection was conducted in a final volume of 1 ml. After 4 hours, fresh media was added and cells were incubated until set times for harvesting, described below, had been reached.
- TUNEL Terminal transferase dUTP Nick-End Labeling
- CFA colony forming assay
- LSC Laser scanning cytometry
- DNase 1 constructs are described in FIGURES 5A and 5B and the used primers in Table 1 .
- the Signal peptide (SP) was removed by deleting the 22 amino acids upstream to the first DNase 1 codon. Partial resistance was rendered to actin by mutating one of the several amino acids involved in binding actin, A1 14F, in DNase 1 constructs C1 1 , C13, C18 and C19.
- the catalytic activity of the enzyme was compromised by mutating one of the two catalytic sites, H252A, in DNase 1 constructs C18 and C19.
- the SV- 40 - Nuclear Localization Signal (NLS), PPKKKRKV was added to the DNase 1 gene at the N-terminus of gene-constructs C07, C13 and C19, and at the C-terminus of gene-constructs C09, C1 1 and C18.
- Klenow Reaction The large fragment of DNA polymerase (Klenow fragment) was used to elongate primer pairs (Integrated DNA Technologies (IDT), Coralville, IA) from an overlapping region where the pairs were annealed to create very long and stable primers to be used in all subsequent mutagenesis reactions except for generating actin-binding and catalytic site mutants.
- IDT Integrated DNA Technologies
- IA Integrated DNA Technologies
- Chloramphenicol transferase (CAT) reporter plasmid (pcDNA/GW- 47/CAT) served as a control (# 12281 -010, Invitrogen).
- GFP Green Fluorescence Protein
- P3-for-20p6 and P6-rc primers generated wiled type DNase-1 (C03) (FIGURES 20, 32).
- SV40 - Nuclear Localization Signal (NLS) was inserted at 5'-prime and 3' prime of C14 to generate constructs C01 and C04, respectively.
- C01 was generated with P1 -NLS-for and P2-NLS-rc primers (FIGURES 20, 31 ) and C04 was generated with P3-NLS-for and P4-NLS-rc primers (FIGURES 20, 32).
- H44A-for/H44A-rc, D53A-for/D53A- rc, Y65W-for/Y65W-rc, V67M-for/V67M-rc, and A1 14R-for/A1 14R-rc primers were generated to replace histidine with alanine at position 44, to replace aspartamine with alanine at position 53, to replace tyrosine with alanine at position 65, to replace valine with alanine at position 67, and to replace alanine with arginine at position 1 14, respectively, in C09.
- constructs (FIGURES 30, 33).
- RNA was extracted from Mel-Juso cells 12 hours, 24 hours, and 36 hours post- transfection using the RNeasy kit (Qiagen, Hilden, Germany) according to the manufacturer's instructions. Quantitative RT-PCR was performed using TaqMan One-Step RT-PCR Master Mix and probes for DNase 1 (Hs00173736_m1 ) and Tubulin (Hs00742828_s1 ) (Applied Biosystems, Foster City, CA) and detected with 7300 Real-Time PCR System (Applied Biosystems). For each sample, 1 g of total RNA was reverse-transcribed at 48 °C for 30 minutes.
- PCR was performed at 95 °C for 10 minutes, followed by 40 cycles of 95 °C for 15 seconds and 60 °C for 1 minute.
- the final reaction volume of 50 ⁇ _ included 1 X Master Mix, MultiScribe (0.25 U/ ⁇ -), RNase Inhibitor Mix (0.4 ⁇ / ⁇ _), 200 nM of DNase 1 construct, or 200 nM (TUBB) forward (ham F) and reverse (ham R) primers.
- the relative levels of gene expression of target mRNA was normalized to endogenous TUBB mRNA using the 2- ⁇ method Experiments were performed three times.
- DNase 1 variant proteins were synthesized using equal amounts (250 ng) of each variant construct plasmid DNA as a template in an in vitro transcription translation kit (PureExpress, New England Biolabs, Ipswich, MA) according to the manufacturer's instructions. As previously demonstrated, DNase 1 activity was determined quantitatively by ELISA (enzyme - linked - immunoabsorbent assay; Orgentec Diagnostika GmbH, Mainz, Germany) according to the manufacturer's instructions. In short, samples were added to a DNA-coated 96 well microplate and incubated for 1 hour at 37 °C.
- HRP horseradish peroxidase conjugated anti-DNase 1 antibodies
- the intensity of the color developed during the enzymatic reaction was measured at OD450 using SPECTRAmax GEMINI XS microplate spectrofluorometer (Molecular Devices, Sunnyvale, CA).
- the color intensity in the ELISA assay reflects the amount of remaining intact DNA substrate after exposure to DNase 1 . As the catalytic activity of a DNase 1 gene construct increases, less DNA substrate remains intact and the colorimetric level is lower. A standard series, as well as positive and negative controls were included in the kit.
- CFA Colony Forming Assay
- Morphological Assessment of Apoptosis Characteristic changes in cell morphology that were used to detect apoptosis include cell membrane blebbing, cytoplasm and nuclear condensation and apoptotic bodies. The morphological changes were visualized by phase contrast microscopy prior to fluorescent microscopy using an Olympus 1 X71 Inverted Microscope (Olympus, Centervalley, PA).
- TUNEL Terminal Transferase dUTP Nick-End Labeling
- LSC Laser Scanning Cytometry for Apoptosis. Twenty-four hours after the cells were transfected with various DNase 1 constructs and controls as described above, the cells were treated with the DeadEnd Fluorometric TUNEL System (Promega) according to the manufacturer's protocol for detection of apoptotic cells. Cells were counterstained with propidium iodide (PI) as previously described. Fluorescein-TUNEL-positive cells were scanned using an X20 objective and their numbers were measured by LSC (CompuCyte Corp., Cambridge, MA, USA).
- Cells were excited by an argon laser at 488 nm and emitted fluorescence was collected through a 530 ⁇ 30 nm band pass filter (green-FITC/TUNEL) and a 625 ⁇ 28 nm band-pass filter (red-PI). Cell population data were analyzed by WinCyte 3.7 (Compucyte Corp.). The cells were plated in triplicates and at least 2,000 cells were measured per sample.
- DNA Fragmentation Assay for Apoptosis Twenty-four hours after the cells were transfected with various DNase 1 constructs and controls as described above, the cells were collected and lysed. Genomic DNA was purified using an Apoptotic DNA Ladder Kit (Roche, Mannheim, Germany) according to the manufacturer's instructions. After 20 minutes of treatment with RNase A (200 Mg/mL; Invitrogen) the samples were analyzed on a 1 % agarose gel supplemented with Syto60 DNA binding dye (Invitrogen, Eugene, OR) and visualized with the Odyssey system (Li-Cor Biosciences, Lincoln, NE).
- RNase A 200 Mg/mL
- Syto60 DNA binding dye Invitrogen, Eugene, OR
- DNase 1 gene construct compositions were generated by fusing an NLS to either the N-terminus or the C-terminus of the DNase 1 gene and by fusing GFP to the C-terminus directly or downstream to the fused NLS.
- mutations were introduced in the DNase 1 gene composition to knockdown DNase 1 protein binding to actin and catalytic activity.
- FIGURES 5A and 5B The resulting various DNase 1 gene constructs are presented in FIGURES 5A and 5B.
- CFA Colony forming assay
- FIGURES 10A-10D Representative examples of characteristic apoptotic morphology and DNA nicking analyzed with phase contrast microscopy and TUNEL assay are presented in FIGURES 10A-10D.
- Substantial numbers of TUNEL positive cells and cells displaying apoptotic characteristics such as cytoplasm and nuclear condensation and cell membrane blebbing were observed only after treatment with actin- resistant DNase 1 gene constructs, C1 1 and C13, and only sporadically, due to background apoptosis, after treatment with other DNase 1 gene constructs, Wild-type DNase 1 and controls (FIGURES 10A-10D).
- Background apoptosis can reflect spontaneous apoptosis that occurs in untreated neoplasms or apoptosis induced by DNA-liposome complexes.
- DNA fragmentation assay for apoptosis DNA fragmentation was most extensive after treating the melanoma cells with the actin-resistant DNase 1 gene constructs (C1 1 and C13), reduced after treatment with either C19 or C07, and minute or absent after treatment with C18, C07 or Wild-type DNase 1 (FIGURE 1 1 ). DNA fragmentation in the form of a smear is characteristic of DNase 1 .
- DNase 1 For inducing the classical DNA laddering form of DNA degradation (low molecular weight DNA fragmentation) DNase 1 requires the removal of DNA binding proteins, which are released during cell damage conditions such as necrosis.
- DNase 1 a genetically modified waste- management nuclease
- the Mel-Juso cell line was chosen due to its high resistance to apoptosis, which makes it a good choice for testing the killing efficiency of pro-apoptotic therapies.
- This cell line is chemoresistant, displays higher resistance to UVB induced apoptosis than other melanoma cell lines, and is not sensitive to Fas or TRAIL induced apoptosis.
- the apoptosis- resistance of Mel-Juso results from multiple molecular changes such as mutations in the pro- apoptotic RAS gene or overexpression of the strong anti-apoptotic Bcl-xL, a member of the Bcl-2 family.
- SP Signal peptide
- NLS Nuclear Localization Signal
- C07 and C09 DNase 1 constructs were generated having the NLS fused to DNase 1 's N-terminus and C-terminus, respectively.
- overexpression of the modified DNase 1 cDNA did not decrease melanoma cell survival (FIGURE 9) nor did it increase the number of cells undergoing apoptosis as assessed by TUNEL assay and measured by LSC (FIGURES 12A- 12B).
- C09 showed no chromatin-degrading effect greater than controls, while the C07 construct displayed a limited chromatin-degrading effect compared to C1 1 and C13.
- the nucleus contains abundant amounts of actin, which is the major natural inhibitor of DNase 1 . Therefore, it was hypothesized that nuclear actin inhibited the activity of the NLS fused DNase 1 constructs, C07 and C09. To test this hypothesis, DNase 1 's binding site (A1 14F) in C07 and C09 was mutated, thus, generating actin-resistant DNase 1 constructs, C13 and C1 1 , respectively. A1 14F substitution is reported to increase the potency of WT DNase 1 by ⁇ 5-fold.
- the generated actin-resistant C1 1 and C13 DNase 1 constructs comprise three genetic alterations: (i) eliminated SP, (ii) fused NLS, and (iii) mutation of actin-binding site.
- C1 1 and C13 significantly decreased melanoma cell survival (FIGURE 9), induced characteristic apoptotic morphological changes (FIGURES 10A-10D), increased the number of cells undergoing apoptosis as assessed by TUNEL assay and measured by LSC (FIGURES 12A-12B), and induced DNA-degradation (FIGURE 1 1 ).
- Quantitated apoptosis assessment (FIGURES 12A-12B) showed an advantage over WT DNase 1 to be significant for C1 1 and only marginally significant for C13.
- constructs having the A1 14F mutation which were only partially resistant to actin inhibition, were extremely efficient inducers of apoptosis.
- gene constructs having incomplete resistance to an inhibitor can nonetheless produce significant apoptosis when directed to the nucleus.
- incomplete resistance can be conferred by knocking out less than all actin-binding sites, or by only partially inhibiting actin binding sites.
- the findings also indicate that the cell-killing efficiency of genetically-modified DNA-degrading enzymes, that have binding sites for inhibitors other than actin, can be increased by engineering suitable mutations of those inhibitor binding sites.
- the killing efficiency of C1 1 and C13 DNase 1 constructs using LSC was assessed, which provides a quantitative analysis of TUNEL positive cells.
- LSC measured a Cat- GFP cell population, representing transfection efficiency, of 10-20%.
- the TUNEL positive cell population after C1 1 construct treatment was 7-21 % and after C13 treatment 4- 8%.
- the ratio of cells undergoing apoptosis to the transfection cells show a killing efficiency of 70-100% for C1 1 and -40% for C13.
- the lower killing efficiency of C13 can reflect lower protein synthesis or reduced catalytic activity due to the N-terminus fusion of the NLS (FIGURE 6B) due to fusion of NLS to the N-terminus of DNase 1 .
- the high killing efficiency of the C1 1 construct indicates that deleting the N- terminus SP and adding a C-terminus NLS to an actin-resistant DNase 1 provided the most effective combination of genetic modifications.
- HSV-tk treatment led to the killing of 9 of 19 rats treated for hepatocellular carcinoma. Therefore, a lack of a 'bystander effect' for the DNase 1 gene therapy, if confirmed by further studies, would provide a substantial safety advantage adding to its high killing efficiency for treatment resistant cancers such as melanoma.
- a waste-management enzyme can be genetically modified to bypass cancer's anti-apoptotic defense mechanism and to trigger apoptosis in human melanoma cells without a pharmacological adjuvant.
- Hr DNase 1 gene constructs and constructs encoding other DNA-degrading enzymes have potential application in vivo application to a variety of treatment-resistant cancers.
- Boone DL Tsang BK., Identification and localization of deoxyribonuclease I in the rat ovary. Biol Reprod 1997, 57:813-21 .
- Linardou H Epenetos AA, Deonarain MP., A recombinant cytotoxic chimera based on mammalian deoxyribonuclease-l. Int J Cancer, 2000, 86:561 -9.
- Polzar B Peitsch MC, Loos R, Tschopp J, Mannherz HG., Overexpression of deoxyribonuclease I (DNase I) transfected into COS-cells: its distribution during apoptotic cell death. Eur J Cell Biol 1993, 62:397-405.
- DNase I deoxyribonuclease I
- DNase-gamma Apoptotic DNA endonuclease gene transfer induces cell death accompanying DNA fragmentation in human glioma cells. J Neuro-Oncol 2003, 63:25-31 .
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Genetics & Genomics (AREA)
- Engineering & Computer Science (AREA)
- Zoology (AREA)
- Molecular Biology (AREA)
- Wood Science & Technology (AREA)
- General Health & Medical Sciences (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- Medicinal Chemistry (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Biotechnology (AREA)
- Microbiology (AREA)
- Immunology (AREA)
- Analytical Chemistry (AREA)
- Biophysics (AREA)
- Biomedical Technology (AREA)
- Pathology (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Hospice & Palliative Care (AREA)
- Epidemiology (AREA)
- Oncology (AREA)
- Toxicology (AREA)
- Gastroenterology & Hepatology (AREA)
- Physics & Mathematics (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
La présente invention concerne une construction génétique comportant un gène effecteur de mort cellulaire programmée comprenant un signal de localisation nucléaire, un peptide signal ayant subi une délétion, un site de liaison résistant aux inhibiteurs, un promoteur, et un activateur. L'invention concerne également un procédé de fabrication d'une construction génétique, par la modification du gène effecteur de mort cellulaire programmée par l'addition d'un signal de localisation nucléaire, la délétion d'un peptide signal, la mutation d'un site de liaison pour un inhibiteur pour le rendre résistant à l'inhibiteur, l'addition d'un promoteur pour une expression exclusive dans des cellules sélectionnées, et l'addition d'un activateur. L'invention concerne en outre un procédé d'élimination de cellules indésirables à partir d'un patient. L'invention concerne également un procédé de traitement du cancer. L'invention concerne également un réseau comportant au moins deux constructions génétiques dans lequel toutes les constructions génétiques sont différentes par rapport au gène effecteur de mort cellulaire programmée et au signal de localisation nucléaire. L'invention concerne également un procédé de personnalisation de traitement du cancer. L'invention concerne également un procédé pour l'accroissement de la résistance de la DNase 1 à la liaison d'actine. L'invention concerne enfin un procédé pour l'accroissement de l'activité catalytique de la liaison DNase 1.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201213392233A | 2012-02-24 | 2012-02-24 | |
US13/392,233 | 2012-02-24 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2013126872A1 true WO2013126872A1 (fr) | 2013-08-29 |
Family
ID=49006294
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2013/027600 WO2013126872A1 (fr) | 2012-02-24 | 2013-02-25 | Stratégie thérapeutique anti-cancéreuse pour combattre la résistance aux traitements contre le cancer et pour permettre la personnalisation de traitement de patients |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2013126872A1 (fr) |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN107677820A (zh) * | 2016-08-02 | 2018-02-09 | 北京金沐医疗科技有限公司 | 一种检测肿瘤抗原的试剂盒及其检测方法 |
CN107677823A (zh) * | 2016-08-02 | 2018-02-09 | 北京金沐医疗科技有限公司 | 一种用于检测肿瘤抗原的蛋白芯片试剂盒及其检测方法 |
CN107677822A (zh) * | 2016-08-02 | 2018-02-09 | 北京金沐医疗科技有限公司 | 检测肿瘤标志物的荧光细胞计数检测试剂盒 |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20090047272A1 (en) * | 2004-04-14 | 2009-02-19 | Appelbaum Jacob G | Compositions with Modified Nucleases Targeted to Viral Nucleic Acids and Methods of Use for Prevention and Treatment of Viral Diseases |
US20100297091A1 (en) * | 2009-05-15 | 2010-11-25 | Wang James J-L | Compositions and methods for treatment of melanomas |
US20120027785A1 (en) * | 2008-03-31 | 2012-02-02 | Dirienzo Joseph M | Chimera comprising bacterial cytotoxin and methods of using the same |
-
2013
- 2013-02-25 WO PCT/US2013/027600 patent/WO2013126872A1/fr active Application Filing
Patent Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20090047272A1 (en) * | 2004-04-14 | 2009-02-19 | Appelbaum Jacob G | Compositions with Modified Nucleases Targeted to Viral Nucleic Acids and Methods of Use for Prevention and Treatment of Viral Diseases |
US20120027785A1 (en) * | 2008-03-31 | 2012-02-02 | Dirienzo Joseph M | Chimera comprising bacterial cytotoxin and methods of using the same |
US20100297091A1 (en) * | 2009-05-15 | 2010-11-25 | Wang James J-L | Compositions and methods for treatment of melanomas |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN107677820A (zh) * | 2016-08-02 | 2018-02-09 | 北京金沐医疗科技有限公司 | 一种检测肿瘤抗原的试剂盒及其检测方法 |
CN107677823A (zh) * | 2016-08-02 | 2018-02-09 | 北京金沐医疗科技有限公司 | 一种用于检测肿瘤抗原的蛋白芯片试剂盒及其检测方法 |
CN107677822A (zh) * | 2016-08-02 | 2018-02-09 | 北京金沐医疗科技有限公司 | 检测肿瘤标志物的荧光细胞计数检测试剂盒 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Los et al. | Apoptin, a tumor-selective killer | |
JP5911481B2 (ja) | 抗癌性融合タンパク質 | |
JP2003513911A (ja) | シスプラチン及び他の薬剤又はリポソーム中に封入された遺伝子を用いてのヒト癌のための療法 | |
JP2006523227A (ja) | Mda−7を含む方法および組成物 | |
US9238682B2 (en) | Anti-cancer therapeutic strategy to overcome cancer resistance and to enable tailoring treatment to patients | |
US20150202250A1 (en) | Anticancer treatment | |
WO2013126872A1 (fr) | Stratégie thérapeutique anti-cancéreuse pour combattre la résistance aux traitements contre le cancer et pour permettre la personnalisation de traitement de patients | |
TW201023898A (en) | Use of a truncated eIF-5A1 polynucleotide to induce apoptosis in cancer cells | |
JP2013173753A (ja) | 多発性骨髄腫細胞を死滅させるためのeIF−5Aの使用 | |
US11773146B2 (en) | Disruption of the WAVE3 protein complex for suppression of invasion and metastasis | |
EP3624825A1 (fr) | Virus oncolytique recombinant | |
EP2797617B1 (fr) | Thérapie d'adjuvant anti-tumeur | |
Lezhnin et al. | Oncotoxic proteins in cancer therapy: Mechanisms of action | |
CN114790225B (zh) | 一种新型内体逃逸肽及其应用 | |
US20080293652A1 (en) | Combination of Ad-P53 and Chemotherapy for the Treatment of Tumours | |
CN114181315B (zh) | 一种内体逃逸肽及其应用 | |
WO2015092756A1 (fr) | Protéine de fusion recombinante prostat et utilisations associées | |
US20090012025A1 (en) | Methods and Compositions for Treatment of Sepsis | |
Yuan et al. | PNAS-4, a novel pro-apoptotic gene, can potentiate antineoplastic effects of cisplatin | |
KR102577502B1 (ko) | 생체분자의 안정적 발현 및 전달을 위한 플라스미드 플랫폼 | |
CN110862446B (zh) | 一种新型肿瘤靶向治疗多肽及其用途 | |
CN116675779B (zh) | 一种靶向Src激酶的短肽及其在系统性真菌感染中的应用 | |
EP3596209B1 (fr) | Système d'enzyme induisant une mort cellulaire sélective | |
KR101715263B1 (ko) | 재조합 hCdc73 단백질 및 이를 포함하는 암 예방 또는 치료용 약학적 조성물 | |
US20130225503A1 (en) | Tex14 peptides as novel antitumor agents |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 13752549 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 13752549 Country of ref document: EP Kind code of ref document: A1 |