This disclosure generally relates to methods of delivering therapeutic compounds to cancer cells.
A number of therapies are currently used for treating cancer, including, for example, chemotherapy, radiation therapy, surgery, gene therapy, and bone marrow transplantation. Therapies that specifically target cancer cells and not non-malignant cells, however, are desirable.
This disclosure describes methods of stimulating macropinocytosis in cancer cells.
In one aspect, a method of stimulating macropinocytosis in cancer cells is provided. Such a method generally includes the steps of contacting the cancer cells with a G-rich nucleic acid that is capable of forming a quadruplex structure to thereby stimulate macropinocytosis in the cancer cells. In certain embodiment, the G-rich nucleic acid is between 10 and 50 nucleotides in length and is greater than 25% G nucleotides. In certain embodiments, the G-rich nucleic acid has a sequence shown in SEQ ID NO: 1. Representative cancer cells include, without limitation, prostate cancer, lung cancer, cervical cancer, breast cancer, colon cancer, pancreatic cancer, renal cell carcinoma, ovarian cancer, leukemia, lymphoma, melanoma, glioblastoma, neuroblastoma, sarcoma, and gastric cancer.
In another aspect, a method of delivering a therapeutic compound to cancer cells is provided. Such a method generally includes the steps of contacting the cancer cells with a G-rich nucleic acid that is capable of forming a quadruplex structure, and contacting the cancer cells with a therapeutic compound. According to this method, the therapeutic compound is taken up (i.e., endocytosed) by the cancer cells via macropinocytosis. In certain embodiment, the G-rich nucleic acid is between 10 and 50 nucleotides in length and is greater than 25% G nucleotides. In certain embodiments, the G-rich nucleic acid has a sequence shown in SEQ ID NO: 1. Representative cancer cells include, without limitation, prostate cancer, lung cancer, cervical cancer, breast cancer, colon cancer, pancreatic cancer, renal cell carcinoma, ovarian cancer, leukemia, lymphoma, melanoma, glioblastoma, neuroblastoma, sarcoma, and gastric cancer.
In certain embodiment, the therapeutic compound is a nucleic acid, a peptide, a small molecule, a drug, a chemical, an antibody or a nanoparticle. Representative nucleic acid, for therapeutic use, include antisense RNA, interfering RNA, immunostimulatory oligonucleotides, triple helix oligonucleotides, transcription factor decoy nucleic acids, aptamers, or plasmid DNA.
In still another aspect, a method of determining whether cancer cells are susceptible or refractory to the antiproliferative effects of a G-rich nucleic acid capable of forming a quadruplex structure is provided. Such a method generally includes the steps of contacting the cancer cells with the G-rich nucleic acid; and determining whether or not macropinocytosis is increased in the cancer cells contacted with the G-rich nucleic acid relative to cancer cells not contacted with the G-rich nucleic acid. Typically, an increase in macropinocytosis by the cancer cells contacted with the G-rich nucleic acid indicates that the cancer cells are susceptible to treatment with the G-rich nucleic acid, while the absence of an increase in macropinocytosis by the cancer cells contacted with the G-rich nucleic acid indicates that the cancer cells are refractory to treatment with the G-rich nucleic acid.
In certain embodiment, the G-rich nucleic acid is between 10 and 50 nucleotides in length and is greater than 25% G nucleotides. In certain embodiments, the G-rich nucleic acid has a sequence shown in SEQ ID NO: 1. Representative cancer cells include, without limitation, prostate cancer, lung cancer, cervical cancer, breast cancer, colon cancer, pancreatic cancer, renal cell carcinoma, ovarian cancer, leukemia, lymphoma, melanoma, glioblastoma, neuroblastoma, sarcoma, and gastric cancer. In some embodiments, the method is performed in vitro with cancer cells obtained from a patient diagnosed with cancer.
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the methods and compositions of matter belong. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the methods and compositions of matter, suitable methods and materials are described below. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety.
This document discloses that G-rich nucleic acids capable of forming quadruplex structures stimulate macropinocytosis in cancer cells but not in non-malignant cells. Macropinocytosis is a type of endocytosis that is distinguishable from other endocytic pathways. Unlike both receptor-mediated endocytosis and phagocytosis, macropinocytosis is not regulated through direct actions of cargo/receptor molecules coordinating the activity and recruitment of specific effector molecules of particular sites at the plasma membrane.
Macropinosomes are derived from actin-rich extensions of the plasma membrane, referred to as ruffles. Membrane ruffling occurs due to actin polymerization near the plasma membrane. As the newly formed actin branch grows, the plasma membrane is forced out, extending the membrane into a ruffle. Macropinosomes are formed when these ruffles fuse back with the plasma membrane and encapsulate a large volume of extracellular fluid in the process. Macropinosome formation can be inhibited with amiloride, an ion exchange inhibitor, or derivatives thereof, with no detectable effect on the other endocytic pathways. Therefore, in concert with the morphological description, suppression with amiloride (and, optionally, elevation in response to growth factor stimulation) is used to define macropinocytosis and distinguish macropinocytosis from other types of endocytosis.
As demonstrated herein, G-rich nucleic acids stimulate micropinocytosis in cancer cells but not in non-malignant cells. G-rich nucleic acids have been shown to adopt intermolecular or intramolecular quadruplex structures that are stabilized by the presence of G-quartets. G-quartets are square planar arrangements of four hydrogen-bonded guanines that are stabilized by monovalent cations. See, for example, Dapic et al. (2003, Nuc. Acids Res., 31:2097-107). Significantly, G-rich nucleic acids have been shown to exhibit antiproliferative effects on a number of different types of cancer cells. See, for example, Bates et al., 2009, Exp. Mol. Path., 86:151-64.
As used herein, G-rich nucleic acids refer to nucleic acids (e.g., DNA or RNA) that contain a guanine content that is sufficient for formation of quadruplex structures. Although there is not a particular guanine content required for quadruplex formation, G-rich oligonucleotides typically are greater than 25% guanine. G-rich nucleic acids include oligonucleotides between, for example, 12 nucleotides and 50 nucleotides in length (e.g., 15, 18, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 33, 35, 38, 40, 42, 45 or 48 nucleotides in length). G-rich nucleic acids also include nucleic acids greater than 50 nucleotides in length including, for example, nucleic acids that are 100 nucleotides or more in length, 250 nucleotides or more in length, 500 nucleotides or more in length, 1000 nucleotides (i.e., 1 kilobase (Kb)) or more in length, 2 Kb or more in length, 3 Kb or more in length, 4 Kb or more in length, or 5 Kb or more in length. G-rich nucleic acids can have modifications to, for example, the backbone (e.g., peptide nucleic acid (PNA), or phosphorothioation), one or more of the bases (e.g., methylation, glycosylation, thiol-modification, or a label (e.g., fluorescence or a radiolabel)), or the 3′ or 5′ end (e.g., a label), provided that the modification does not disrupt the ability of the G-rich nucleic acid to form quadruplex structures.
Because macropinocytosis in cancer cells is stimulated by G-rich nucleic acids, this phenomenon can be utilized to deliver one or more therapeutic compounds to the cancer cells. A therapeutic compound that can be delivered to cancer cells includes, without limitation, nucleic acids, peptides, small molecules, drugs, chemicals, antibodies or nanoparticles. Since non-malignant cells still undergo macropinocytosis to a limited degree, the specificity afforded by using therapeutic compounds such as nucleic acids may be preferred. Representative nucleic acids can be, for example, antisense RNA, interfering RNA (e.g., siRNA), immunostimulatory oligonucleotides (e.g., CpG motif-containing oligonucleotides), triple helix oligonucleotides, transcription factor decoy nucleic acids, aptamers, or plasmid DNA. In addition, a therapeutic compound such as a nucleic acid may be linked to or contiguous with the G-rich nucleic acid.
One or more G-rich nucleic acids and/or one or more therapeutic compounds can be delivered to cancer cells via any number of means. For example, one or more G-rich nucleic acids and/or one or more therapeutic compounds can be delivered to cancer cells via direct injection (e.g., into a solid tumor), intravenous administration, intraperitoneal administration, subcutaneous administration, oral administration or administration by inhalation. The one or more G-rich nucleic acids can be delivered to the cancer cells prior to delivery of the one or more therapeutic compounds (e.g., to allow the induction of macropinocytosis to occur), or the one or more G-rich nucleic acids and the one or more therapeutic compounds can be delivered to cancer cells simultaneously or essentially simultaneously. If delivered simultaneously, the one or more G-rich nucleic acids and the one or more therapeutic compounds can be delivered via a single composition or via separate compositions.
G-rich nucleic acids have been shown herein to stimulate macropinocytosis in prostate cancer, lung cancer, cervical cancer and breast cancer. Since, in addition to prostate cancer, lung cancer, cervical cancer and breast cancer, G-rich nucleic acids have been shown to exhibit antiproliferative effects against colon cancer, pancreatic cancer, renal cell carcinoma, ovarian cancer, leukemia and lymphoma, melanoma, glioblastoma, neuroblastoma, sarcoma, and gastric cancer, it is expected that G-rich nucleic acids would stimulate macropinocytosis in these cancers as well.
Whether or not macropinocytosis is stimulated can be used as a marker to determine whether cancer cells are susceptible or refractory to the antiproliferative effects of a G-rich nucleic acid. For example, cancer cells treated with a G-rich nucleic acid can be evaluated to determine whether or not there is an increase in macropinocytosis. An increase in macropinocytosis in cancer cells treated with a G-rich nucleic acid generally indicates cancer cells that are susceptible to the G-rich nucleic acid, while the lack of an increase indicated cancer cells that are refractory to the G-rich nucleic acid.
In accordance with the present invention, there may be employed conventional molecular biology, microbiology, biochemical, and recombinant DNA techniques within the skill of the art. Such techniques are explained fully in the literature. The invention will be further described in the following examples, which do not limit the scope of the methods and compositions of matter described in the claims.
Oligodeoxynucleotides were purchased from Invitrogen (Carlsbad, Calif.). Sequences used for this study include: AS1411, 5′-d(GGT GGT GGT GGT TGT GGT GGT GGT GG) (SEQ ID NO:1); FL-AS1411 (fluorophore-labeled AS1411), 5′-Fluor-d(TTT GGT GGT GGT GGT TGT GGT GGT GGT GG) (SEQ ID NO:2), where Fluor is either 5-Carboxyfluorescein (FAM, used for flow cytometry studies) or Alexa Fluor 488 (used for confocal microscopy); tAS1411, 5′-d(TTT GGT GGT GGT GGT TGT GGT GGT GGT GG) (SEQ ID NO:3); FL-CRO, 5′-Fluor-d(TTT CCT CCT CCT CCT TCT CCT CCT CCT CC) (SEQ ID NO:4); CRO, 5′-d(CCT CCT CCT CCT TCT CCT CCT CCT CC) (SEQ ID NO:5); and tCRO, 5′-d(TTT CCT CCT CCT CCT TCT CCT CCT CCT CC) (SEQ ID NO:6). Unmodified oligonucleotides were purchased in the desalted form, whereas fluorescently labeled sequences were HPLC purified. The 29-mer sequences were used for some experiments because quenching of the fluorophore occurred when it was located adjacent at the 5′-terminal base of the AS1411 sequence, so a spacer consisting of 3 thymidines was added. The antiproliferative activities of 29-mer sequences, with and without the fluorophore, were comparable to the synthesized 26-mer AS1411 sequence, as well as to AS1411 obtained from Antisoma (see
All cells were obtained from the American Type Culture Collection (ATCC) and grown in a humidified incubator maintained at 37° C. with 5% CO2. Hs27 (non-malignant human foreskin fibroblasts), DU145 (hormone-refractory prostate cancer), A549 (non-small cell lung cancer), HeLa (cervical adenocarcinoma), MCF-7 (hormone-dependent breast cancer) and MDA-MB-231 (hormone-independent breast cancer) cells were grown in DMEM supplemented with 10% fetal bovine serum (FBS; Life Technologies), 62.5 μg/mL penicillin and 100 μg/mL streptomycin (Hyclone Laboratories, Logan, Utah). MCF-10A cells (immortalized human breast epithelial cells) were grown in MEBM supplemented with all the components of MEGM bullet kit (Lonza, Allendale, N.J., Catalog No. 3150) except for the GA-1000. Cells were plated at 50% confluence and incubated 18 h to allow adherence, and then the medium was changed for fresh supplemented medium and treated by addition of oligodeoxynucleotides directly to the culture medium to give the final concentration indicated in the Description of the Drawings. Dynasore and cytochalasin D were dissolved in DMSO. Amiloride was dissolved in serum-free medium. Cells were pre-treated with inhibitors in serum-free medium for either 30 min (cytochalasin D) or 60 min (dynasore and amiloride). Cells for biochemical analyses were lysed in lysis buffer (150 mM NaCl, 2 mM EDTA, 50 mM Tris-HCl, 0.25% deoxycholic acid, 1% IGEPAL® CA-630, pH 7.5) containing protease and phosphatase inhibitor cocktails (Calbiochem, Catalogs No. 539134 and 544625) for 20 min at 4° C. and then cleared by centrifugation at 16,000×g for 10 min at 4° C. All protein concentrations were determined using the BCA assay (Pierce, Rockford, Ill.).
To analyze uptake of the oligodeoxynucleotides or dextran-10K (macropinocytic marker) by flow cytometry, 2×105 cells in fresh supplemented culture medium (2.5 ml) were plated into 6-well plates for 18 h. After complete adhesion, the cells were incubated with 5′-FAM tagged oligodeoxynucleotides or Alexa Fluor 488 tagged dextran-10K and incubated as indicated in the Description of the Drawings. Cells were washed once with ice-cold PBS, incubated with 1 μg/ml 7-amino-actinomycin D (7-AAD) for 5 min on ice or 1 μg/ml propidium iodide (PI), and washed twice with ice-cold PBS. Cells were then treated with 0.01% trypsin/0.5 mM EDTA (300 μl) for 3 min prior addition 3 ml supplemented culture medium. The cells were then centrifuged and resuspended in 0.5 ml of 1% paraformaldehyde for analysis by flow cytometry using a FACScalibur cytometer (BD Biosciences, Mountain View, Calif.).
Cells (4×104) in fresh supplemented culture medium were plated on 18 mm diameter glass cover slips for 18 h. The media was removed and replaced with serum-free medium containing 10 μM oligodeoxynucleotide, dextran-10K, or transferrin and incubated as describe in the Description of the Drawings. After incubation, cells were washed 3 times with ice-cold PBS, fixed in 4% paraformaldehyde in PBS for 30 min at room temperature, and washed three times with PBS. After washing, the cover slips were mounted on glass slides with ProLong Antifade (Molecular Probes) according to the manufacturer's directions to inhibit photobleaching. Immunofluorescence was documented with an LSM 510 inverted confocal laser-scanning microscope (Carl Zeiss, Oberkochen, Germany) equipped with an Omnichrome argon-krypton laser. Images were obtained with a Zeiss Plan-Apo 63× oil immersion objective (1.4 NA).
Plated cells were washed three times with ice-cold PBS and added freshly prepared solution of 0.5 mg/ml of a cell-impermeable biotinylating agent (sulfo-NHS-biotin, Pierce, Rockford, Ill.) in PBS. After incubation for 30 min at 4° C., cell were washed once with ice-cold TBS (50 mM Tris-HCl, 150 mM NaCl, pH 7.5), incubated with ice-cold supplemented culture media for 10 min at 4° C., and then washed twice with TBS. Biotinylated proteins were precipitated by incubating with high capacity Neutravidin agarose (Pierce) for 2 h at 4° C. with gentle agitation, and then washed with ice-cold lysis buffer.
The nucleolin siRNA sequences were: 5′-GGU CGU CAU ACC UCA GAA Gtt/5′-CUU CUG AGG UAU GAC GAC Ctc (NCL1) (SEQ ID NO:7); 5′-GGC AAA GCA UUG GUA GCA Att/5′-UUG CAU CCA AUG CUU UGC Ctc (NCL2) (SEQ ID NO:8); and 5′-CGG UGA AAU UGA UGG AAA Utt/5′-AUU UCC AUC AAU UUC ACC Gtc (NCL3) (SEQ ID NO:9), targeted to non-conserved regions of the nucleolin open reading frame (GenBank Accession No. NM—005381). BLAST analysis showed no homology of the siRNA sequences to any other sequence in the Human Genome Database. The siRNA nucleotides were chemically synthesized and annealed by Ambion Inc. (Austin, Tex.). Nucleolin siRNAs (30 nM) were transfected in DU145 cells using Lipofectamine 2000 (Invitrogen), according to the manufacturer's directions. The scrambled siRNA used as a negative control was obtained from Ambion.
Samples were resolved by 10% SDS-Tris polyacrylamide gel electrophoresis and then electrotransferred onto polyvinylidine fluoride (PVDF) membranes (Millipore, Bedford, Mass.) in Tris-glycine buffer containing 20% methanol. Proteins were detected by immunoblotting as described (Reyes-Reyes et al., 2006, Exp. Cell Res., 312:4056-69). In some cases, PVDF membranes were stripped of bound antibodies using 62.5 mM Tris-HCl, pH 6.7, 100 mM 2-mercaptoethanol, 2% SDS for 30 min at 60° C. and then reprobed as described in the Description of the Drawings.
In some experiments, densitometry was used to measure band intensities by scanning autoradiographic films and using UN-SCAN-IT gel software (Silk Scientific Corporation). Band intensities were normalized as indicated in the Description of the Drawings. The statistical comparisons between AS1411-treated and control groups were carried out using Student's t test, and differences are indicated as *(p<0.05).
To first identify suitable conditions to study the mechanism of AS1411 uptake, the timing and serum-dependence of uptake was analyzed in DU145 prostate cancer cells, which are sensitive to AS1411. Uptake of FL-AS1411, a fluorescently labeled version of the active aptamer, and FL-CRO, a fluorescently labeled control oligonucleotide with no antiproliferative activity, was examined by flow cytometry with gating to exclude non-viable cells. Cell-associated fluorescence was not influenced by washing the cells with dextran sulfate to remove the extracellular fluorophore-labeled DNA or by adding trypan blue to quench external fluorescent signals prior to flow cytometry, ruling out the possibility that fluorophore-labeled DNA fluorescent signal is emanating from cell surface (see
FL-AS1411 uptake was detected as early as 5 min, with maximum uptake between 2 h and 4 h, and decreasing after 8 h under these conditions (
To determine whether AS1411 uptake occurs through an active uptake process, the temperature-dependence of AS1411 uptake in cancer cells (DU145, HeLa, MDA-MB-231) and non-malignant Hs27 skin fibroblasts was evaluated using flow cytometry. In all cell types, the uptake of FL-AS1411 and FL-CRO showed strong temperature dependence. However, in contrast to the original hypothesis, Hs27 cells appeared to have a higher uptake of AS1411 than any of the cancer cells analyzed (
To confirm that uptake of AS1411 occurs by endocytosis, the involvement of the actin cytoskeleton, which has been implicated in regulating endocytic pathways, was evaluated. To this end, DU145 and Hs27 cells were pre-treated with an actin polymerization inhibitor. 5 μM cytochalasin D, and assessed for FL-AS1411 uptake by flow cytometry. Cytocholasin D-treated cells showed a decrease in FL-AS1411 uptake compared with the untreated cells (
Recent work has showed that internalization of DNA plasmids or oligonucleotides can be mediated through macropinocytosis (Basner-Tschakarjan et al., 2004, Gene Ther., 11:765-74; Fumoto et al., 2009, Mol. Pharm., 6:1170-9; Wittrup et al., 2007, J. Biol. Chem., 282:27897-904), an actin-driven, ligand-independent mechanism in which cells “gulp” the surrounding medium and any macromolecules it contains. This endocytic mechanism has been shown to be sensitive to amiloride, a specific inhibitor of Na+/H exchange (West et al., 1989, J. Cell. Biol., 109:2731-9) and, therefore, the effect of this inhibitor on FL-AS1411 uptake was tested. It was found that amiloride pre-treatment caused a reduction in FL-AS1411 uptake only in DU145 cancer cells, but not in the non-malignant Hs27 cells (
Further experiments were performed to confirm the identity of the vesicles containing FL-AS1411. Macropinosomes lack a clathrin coat and can be distinguished from endosomes by their comparative inability to concentrate receptors (Thomas et al., 2004, PLoS Biol., 2:1363-80). Therefore, cells were incubated with dextran-Alexa Fluor 488 together with a ligand for the transferrin receptor, transferrrin-Alexa Fluor 594 (
AS1411 causes a change in cancer cell morphology that is characterized by vacuolization, irregular nuclei, and swollen cells (Xu et al., 2001, J. Biol. Chem., 276:43221-30). Therefore, the effect of AS1411 on macropinocytosis in DU145 cells and non-malignant Hs27 cells was investigated. Flow cytometry experiments indicated a significant increment in the uptake of the macropinocytic marker, dextran, in DU145 cells treated with tAS1411 (which is FL-AS1411 without the fluorescent label) for 24, 48, or 72 h (
It has been shown previously that nucleolin is the primary molecular target of AS1411 (Bates et al., 2009, Exp. Mol. Pathol., 86:151-64), and it was originally hypothesized that surface nucleolin may serve as a receptor for AS1411. However, the data presented herein are not consistent with that hypothesis because they indicate that uptake occurs, not by classical receptor-mediated endocytosis, but by macropinocytosis. Therefore, the role nucleolin plays in AS1411 uptake was evaluated. The effect of anti-nucleolin mAbs on uptake of FL-AS1411 was first assessed after 2 h incubation using flow cytometry and it was found that none of the anti-nucleolin mAbs tested affected uptake of FL-AS1411 (
The results shown in
A number of additional G-rich oligonucleotides were obtained and used to evaluate whether or not macropinocytosis was increased in cancer cells using the methodology described herein. For example, the following sequences were used:
A small number of the G-rich sequences evaluated did not stimulate macropinocytosis, but most of the G-rich oligonucleotides used increased macropinocytosis in DU145 prostate cancer cells from 10% over the untreated control up to 51% over the untreated control cells.
In addition, the G-rich oligonucleotides disclosed in Dapic et al. (2003, Nuc. Acids Res.,31:2097 -107; KS-A though KS-I) and the G-rich oligonucleotides (e.g., telomere homologs, GT oligonucleotides, Stat3 binders, Dz13, and triplex oligonucleotides with aptameric effects) disclosed in Bates et al. (2009, Exp. Mol. Path., 86:151-64) and references therein are shown to stimulate macropinocytosis in cancer cells.
It has recently been discovered that AS1411 can stimulate macropinocytosis (MP) in cancer cells and this finding was verified by several different methods and in multiple cancer cell lines (Table 1).
DU145 s.c. xenografts are established on the rear flanks of 6-week old male athymic (nu/nu) mice. When the tumors reach approximately 400 mm3, mice are treated by i.p. injections of AS1411 twice daily for 7 days at a dose of 10 mg/kg/dose. Following euthanasia of mice, tumors are excised, fixed in formalin, and processed for transmission electron microscopy (TEM), standard histochemical staining (H&E, PAS) and immunohistochemistry. Tumor cell morphology is evaluated, the presence of macrophages and other immune cells is assessed, and markers of various forms are stained for cell death and molecules that are involved in MP and methuosis (Ras, Rac1, etc.). To visualize MP in vivo, a protocol similar to that first described by Lencer et al. (1990, Am. J. Physiol., 258:C309-17) is used. Briefly, this involves intravenous infusion of fluorophore-labeled 10 kDa fixable dextran (a fluid phase marker, which was used for the cell-based studies), followed by in vivo fixation by perfusion with a paraformaldehyde/lysine/periodate solution. Post-mortem tissues are flash frozen and cut into semi-thin sections using a microtome. Specimens then are observed by fluorescence microscopy and TEM (following photochemical reaction of p-diaminobenzidine, catalyzed by the fluorophore). To examine the role of MP in initial uptake and antitumor activity of AS1411, amiloride, a Na+/H+ exchanger inhibitor that blocks MP will be utilized. Amiloride is FDA-approved for human use as a diuretic and has been used extensively in experimental animals, including as an in vivo inhibitor of MP. To examine initial uptake, mice are co-injected with 10 mg/kg fluorophore-labeled AS1411 plus 150 μg amiloride, then mice are euthanized after 2 h and tumors excised, fixed and examined by fluorescence microscopy. As a control for specificity, the effect of amiloride also is assessed on uptake of fluorophore-labeled transferrin (which is internalized by receptor-mediated endocytosis and not MP) using in vivo dosing that has been described for other purposes (Sparks et al., 1983, Cancer Res., 43:73-7). It will be examined whether daily amiloride co-treatment can block AS1411 anti-tumor activity (assessed by tumor volume) using proper controls to account for any effects of amiloride alone on tumor growth.
A lack of apoptosis in DU145 cells treated with AS1411 is confirmed using the methods outlined below for U937 cells (
As described above, it is known that AS1411 can induce an unusual form of cell death in cancer cells. It was previously shown that G-rich oligonuclotides could induce cell death selectively in cancer cells compared to non-malignant cells, but it was noted that the morphology of the cells was inconsistent with death by apoptosis. The timing of cell death was also quite unusual, with continuous exposure (at 10 μM AS1411) for 7 days or more required to cause complete cell death for most cancer cells tested. Interestingly, this time course is similar to that seen during induction of methuosis by ectopic Ras expression. However, cell death was also dose-dependent and occurred within hours in DU145 cells treated with 40 μM AS1411. Based on the various published reports, the cell death mechanism in U937 leukemia cells was investigated, and it was confirmed that cell death was not by apoptosis (
It also appears unlikely that AS1411-induced cell death is due to autophagy. Not only is the ultrastructural morphology quite different (the vacuoles in AS1411-treated cells have single membranes and do not usually contain organelles), but also the autophagy inhibitor, 3-methyladenine (3-MA), did not inhibit AS1411 activity (
Protocols were recently established for the growth of DU145 cells as spheroids using low adherence plates and specialized medium, and experiments showed that AS1411 can cause disintegration of spheroids (
Expression of EGFR, Ras, and Rac pathways was evaluated at various times following treatment of DU145 cells with AS1411 or controls. Total protein levels for EGFR, H/K/N-Ras, and Rac 1/2/3 is determined. Constitutive and EGF-stimulated activation of EGFR receptor is examined by looking at receptor phosphorylation, dimerization and degradation in the absence or presence of AS1411. Ras and Rac activation is assessed using binding domain pull-downs (Raf-RBD and PAK-PBD) followed by Western blotting for various isoforms. Activation of downstream pathways is determined by Western blotting for phosphorylated forms of ERK, Akt, and p38MAPK. Methods for all of these assays are well established and routinely used. For any of the downstream pathways that are activated, it will also be determined whether or not they are essential for AS1411 activity by using siRNA knockdown and pharmacological inhibitors. AS1411 activity is evaluated based on the stimulation of MP, percentage of cells with vacuolization, and anti-proliferative activity (where possible, because persistent inhibition of some targets will be toxic). Next, the effects of constitutively active (CA) or dominant negative (DN) forms of Ras and Rac1 are examined on AS1411-induced MP and cell vacuolization (and, where possible, cell death). In addition, the EGFR-dependence of AS1411-stimulared MP is confirmed using siRNAs to knockdown EGFR expression. To investigate possible roles of nucleolin in mediating upstream events during the AS1411-induced activation of Rac (
AS1411-induced MPsomes is characterized and it is confirmed that they undergo abnormal trafficking, as observed during Ras-induced methuosis. Evidence that AS1411-induced MPsomes avoid lysosomal fusion also is relevant. Co-localization of AS1411-induced MPsomes is evaluated with markers for various endosomes and lysosomes (e.g., EEA1, LAMP1, Lysotracker Red, Magic Red, acridine orange, Rab5, Rab7). Changes in lipid composition during trafficking of the AS1411-induced vesicles is probed by expression of GFP-2xFYVE, which specifically binds PtdIns(3)P. The studies for MPsome trafficking are carried out in live cells and are tracked over time using time-lapse video microscopy (both standard and confocal). Additionally, the role of Arf6 and GIT1 in mediating AS1411 effects is examined. These factors lie downstream of Rac, are important for MPsome trafficking, and were recently found to play a role in methuosis.
Finally, it will be determined whether the AS1411-induced molecular changes found in cultured cells also occur in vivo. This is achieved by immunohistochemical staining of AS1411-treated tumors to detect altered protein levels or localization.
The ability of AS1411 pre-treatment to improve delivery and activity of molecules that cannot enter cells by passive diffusion is evaluated. These will include siRNAs to polo-like kinase (PLK1), a DNA plasmid encoding the luciferase reporter gene, an antibody to PLK1, phalloidin (a cell-impermeable toxin targeting actin), and gelonin (a cell impermeable toxin that inactivates ribosomes). These examples were chosen because methods for their use (including dosing) have been previously reported and because, in some cases, they have demonstrated activity against prostate cancer cells when delivered in a targeted fashion. Delivery is monitored by flow cytometry and confocal microscopy using fluorescently tagged molecules (siRNAs, plasmids), by indirect immunofluorescence (gelonin, PLK1 antibody), or by the intrinsic fluorescence of the molecule (phalloidin). Functional outputs include target knockdown, luciferase activity, and antiproliferative effects measured using the MTT assay after 4 and 7 days of treatment. For the last assay, the combination index for agents (added at the same time or 48 h after AS1411) is determined to identify any synergistic or additive effects. Effects on non-malignant cells, including dendritic cells and macrophages, are assessed. Similar methods are used to test AS1411 in combination with agents that activate MP and Rac. These include EGF, TAT protein transduction domain (a cell penetrating peptide), caffeine, hyaluronan, methamphetamine, and FTY720 (a sphingosine-1-phosphate receptor agonist). These were chosen because methods for their use are well established and their ability to stimulate MP or Rac activation has been well documented. In addition, many of these are FDA-approved for human use in non-cancer indications (EGF, methamphetamine, hyaluronan, FTY720, caffeine).
Pre-treatment of cancer cells with AS1411 is used to increase cellular delivery of molecules that do not easily cross the plasma membrane. Furthermore, due to the unique properties of MPsomes, delivery by MP leads to increased functional activity. Thus, treatment with AS1411, followed by administration of an anticancer siRNA, for example, leads to a synergistic increase in anticancer effects without harming normal cells. Another strategy to potentiate the effects of AS1411 is to combine it with agents that promote MP and activation of Rac. This leads to increased macropinocytic uptake of AS1411 and/or enhanced methuosis. It has already been shown that pre-treatment of cancer cells with AS1411 leads to induced uptake of dextran, AS1411, or transferrin (by MP) from the culture medium. In addition, the uptake of fluorescently labeled duplex siRNA in DU145 cells pre-treated with AS1411 (10 μM, 48 h) was examined, and a substantial increase in siRNA delivery was observed (
It is to be understood that, while the methods and compositions of matter have been described herein in conjunction with a number of different aspects, the foregoing description of the various aspects is intended to illustrate and not limit the scope of the methods and compositions of matter. Other aspects, advantages, and modifications are within the scope of the following claims.
Disclosed are methods and compositions that can be used for, can be used in conjunction with, can be used in preparation for, or are products of the disclosed methods and compositions. These and other materials are disclosed herein, and it is understood that combinations, subsets, interactions, groups, etc. of these methods and compositions are disclosed. That is, while specific reference to each various individual and collective combinations and permutations of these compositions and methods may not be explicitly disclosed, each is specifically contemplated and described herein. For example, if a particular composition of matter or a particular method is disclosed and discussed and a number of compositions or methods are discussed, each and every combination and permutation of the compositions and the methods are specifically contemplated unless specifically indicated to the contrary. Likewise, any subset or combination of these is also specifically contemplated and disclosed.
This invention was made with government support under Grant No. CA 122383 awarded by the National Institutes of Health. The government has certain rights in the invention.
Filing Document | Filing Date | Country | Kind | 371c Date |
---|---|---|---|---|
PCT/US11/27124 | 3/4/2011 | WO | 00 | 8/23/2012 |
Number | Date | Country | |
---|---|---|---|
61310419 | Mar 2010 | US |