WO2018019284A1 - Compounds for enhancing bax/bcl-2 expression and activity and therapeutic use thereof - Google Patents
Compounds for enhancing bax/bcl-2 expression and activity and therapeutic use thereof Download PDFInfo
- Publication number
- WO2018019284A1 WO2018019284A1 PCT/CN2017/094855 CN2017094855W WO2018019284A1 WO 2018019284 A1 WO2018019284 A1 WO 2018019284A1 CN 2017094855 W CN2017094855 W CN 2017094855W WO 2018019284 A1 WO2018019284 A1 WO 2018019284A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cancer
- bcl
- dipyridamole
- cell
- bax
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Ceased
Links
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/519—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/435—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
- A61K31/4353—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
- A61K31/436—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having oxygen as a ring hetero atom, e.g. rapamycin
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/513—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7042—Compounds having saccharide radicals and heterocyclic rings
- A61K31/7052—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
- A61K31/706—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
- A61K31/7064—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
- A61K31/7068—Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
Definitions
- the invention provides a method of enhancing the expression and activity of BAX/BCL-2 and related therapeutic use.
- the BCL-2 protein family determines the commitment of cells to apoptosis, an ancient cell suicide programme that is essential for development, tissue homeostasis and immunity. These proteins govern mitochondrial outer membrane permeabilization (MOMP) and can be either pro-apoptotic (Bax, BAD, Bak and Bok among others) or anti-apoptotic (including Bcl-2 proper, Bcl-xL, and Bcl-w, among an assortment of others) .
- MOMP mitochondrial outer membrane permeabilization
- Apoptosis manifests in two major execution programs downstream of the death signal: the caspase pathway and organelle dysfunction, of which mitochondrial dysfunction is the best characterized (for reviews, see Green and Reed 1998; Thornberry and Lazebnik 1998) .
- the caspase pathway As the BCL-2 family members reside upstream of irreversible cellular damage and focus much of their efforts at the level of mitochondria, they play a pivotal role in deciding whether a cell will live or die.
- Apoptosis can be initiated via two pathways, the extrinsic and the intrinsic pathway. Whereas the extrinsic pathway is initiated through stimulation of cell surface death receptors, the intrinsic pathway is activated by many different intracellular stresses and is elicited by most chemotherapy agents.
- the BCL-2 family proteins are predominantly involved in the intrinsic pathway, in which they regulate mitochondrial outer membrane permeabilization (MOMP) .
- MOMP results in the release of apoptosis-triggering factors, such as cytochrome c, from the mitochondrial intermembrane space into the cytoplasm in which they activate a cascade of caspases that execute widespread proteolytic events leading to cellular demise (3) .
- MOMP marks commitment to death and is often considered to be the point of no return for a cell.
- Bcl-2 Overexpression of Bcl-2 was subsequently described in prostate, breast and colon carcinomas, as well as glioblastomas. Overexpression of Mcl-1, another antiapoptotic, Bcl-2-related protein, was identified in relapsed acute myelogenous leukemia, and was associated with poor prognosis. Other changes in Bcl-2-related protein expression identified in cancer cells include different mutations in the Bax gene, and changes in the proapoptotic to antiapoptotic Bcl-2 protein ratio (Bettaieb et al., 2003; and Oncogene (2004) 23, 2934–2949) .
- Bcl-2 and related antiapoptotic proteins have been demonstrated to inhibit cell death induced by many stimuli, including growth factor deprivation, hypoxia and oxidative stress.
- many stimuli including growth factor deprivation, hypoxia and oxidative stress.
- antiapoptotic Bcl-2-family proteins to suppress cell death induced by cytotoxic anticancer drugs that makes these proteins particularly interesting as potential targets for cancer drug discovery.
- Bcl-2 operates at a distal point in a conserved cell death pathway utilized by most anticancer drugs, constituting a form of intrinsic chemoresistance, distinct from the previously identified mechanisms involving drug efflux, drug metabolism, drug inactivation and related mechanisms. This observation presumably explains why expression of a variety of Bcl-2-family proteins has been shown to be of prognostic significance for many types of cancer and leukemia treated by chemotherapy.
- Defects in the expression of proapoptotic members of the BCL-2 family also occur in cancer, resulting in loss of the tumor suppressor function of these killer genes.
- the best documented is BAX, where homozygous deletions or inactivating mutations have been identified, particularly in cancers that arise with microsatellite instability because of defective DNA mismatch repair.
- the human BAX gene contains a homopolymeric stretch of eight guanosine residues in the sense strand that is a target for frame-shift mutations.
- Defective expression of proapoptotic BCL-2-family genes also occurs in the setting of loss of p53 function.
- p53 transcription factor Among the direct targets of the p53 transcription factor are BAX, BID, PUMA and NOXA, thus demonstrating strong connections between genome surveillance by p53 and cell death genes of the BCL-2 family. More recently, cytosolic interactions of p53 protein with pro-and antiapoptotic Bcl-2-family proteins have been observed, directly modulating the bioactivities of the p21-Bax and p26-Bcl-2 proteins, and suggesting that p53 regulates the Bcl-2 family at both transcriptional and post-transcriptional levels. The activities of additional proapoptotic members of the Bcl-2 family are also suppressed through post-translational modifications.
- proapoptotic protein BAD is phosphorylated by Akt (PKB) and other protein kinases known to be hyperactive in cancers, resulting in its sequestration by 14-3-3 (Oncogene (2008) 27, 6398–6406) .
- PAkt Akt
- 14-3-3 Oncogene (2008) 27, 6398–6406
- Bcl-2 prevents apoptosis induced by most and chemotherapeutic drugs, including alkylating agents, and topoisomerase inhibitors.
- fibroblasts deficient in both Bax and Bak are resistant to apoptosis induced by various agents that induce mitochondrial stress.
- fibroblasts with single deficiency for either Bax or Bak are not significantly defective for apoptosis induced by these agents, suggesting that both Bax and Bak proteins may be functionally redundant. This apparent redundancy may be cell type-dependent as Bax-deficient HCT116 cells are resistant to apoptosis induced by staurosporine (Theodorakis et al., 2002) .
- a number of anticancer agents can directly permeabilize the outer membrane of mitochondria, at least in vitro, and overexpression of Bcl-2 prevents arsenic-and lonidamine-induced mitochondrial permeabilization and apoptosis.
- Bcl-2 proteins Based on the role of Bcl-2 proteins in regulating mitochondrialmembrane permeabilization, and their frequently modified expression in human cancers, Bcl-2 proteins are legitimate targets (antiapoptotic members) or prototypes (proapoptotic members) for inducing apoptosis either by themselves or in association with other anticancer drugs.
- molecular markers should be used when using Bcl-2-targeted therapies since not all the apoptosis-resistant cells overexpress Bcl-2, as demonstrated in the breast cancer cell lines from the NCI cell screen (Nieves-Neira and Pommier, 1999; and Oncogene (2004) 23, 2934–2949) .
- the Bcl-2 protein is a suppressor of programmed cell death that homodimerizes with itself and forms heterodimers with a homologous protein Bax, a promoter of cell death.
- Bax/Bcl-2 ratio can act as a rheostat which determines cell susceptibility to apoptosis.
- the subcellular location of some Bcl-2 family members also appears to regulate their function.
- Bcl-2 and a fraction of Bcl-xL can be found on the mitochondrial outer membrane.
- Bax exists predominantly in the cytosol before apoptosis induction. Early during apoptosis, Bax translocates from the cytosol to mitochondria where it participates in mitochondrial disruption and the release of cytochrome c.
- Modulating the insertion step can regulate apoptosis.
- Lower levels of this ratio may lead to resistance of human cancer cells to apoptosis.
- Bax/Bcl-2 ratio can affect tumor progression and aggressiveness (Iran Biomed J. 2015 Apr; 19 (2) : 69–75) .
- Bcl-2 inhibitors in different phases of human clinical trials such as AT-101 (R- (-) -gossypol) , ABT-199 (venetoclax) , ABT-737, ABT-263 (navitoclax, orally available derivative of ABT-737) , GX15-070 (obatoclax) and TW37.
- AT-101 R- (-) -gossypol
- ABT-199 venetoclax
- ABT-737 ABT-263
- GX15-070 obatoclax
- Bcl-2 via its BH4 domain, cooperates with numerous proteins regulating different cellular pathways involved in tumor progression and chemoresistance such as hypoxia and angiogenesis.
- a small molecule namely BDA-366 was discovered as a potent and effective BH4 domain antagonist, displaying remarkable anticancer activity in vitro and in vivo, thus providing the proof-of-concept of this approach.
- Another innovative approach to inhibit Bcl-2 comes from the evidence that human bcl-2 gene contains a GC-rich sequence located in its promoter with the potential to form G-quadruplex structures and functions as a transcriptional repressor element. Therefore, G-quadruplex-specific ligands can regulate the transcription of bcl-2 through stabilization of quadruplex structure (AGING, April 2016, Vol. 8 No. 4) .
- Bcl-XL is a major anti-apoptotic protein in the Bcl-2 family whose overexpression is more widely observed in human lung cancer cells than that of Bcl-2, suggesting that Bcl-XL is more biologically relevant and therefore a better therapeutic target for lung cancer.
- Bcl-XL is a major anti-apoptotic protein in the Bcl-2 family whose overexpression is more widely observed in human lung cancer cells than that of Bcl-2, suggesting that Bcl-XL is more biologically relevant and therefore a better therapeutic target for lung cancer.
- Bcl-XL Two new Bcl-XL inhibitors (BXI-61 and BXI-72) that exhibit selective toxicity against lung cancer cells were compared with normal human bronchial epithelial cells.
- BXI-61 and BXI-72 preferentially bind to Bcl-XL protein but not Bcl2, Bcl-w, Bfl-1/A1 or Mcl-1 in vitro with high binding affinities.
- Treatment of cells with BXI-72 results in disruption of Bcl-XL/Bak or Bcl-XL/Bax interaction, oligomerization of Bak and cytochrome c release from mitochondria.
- BXI-61 and BXI-72 exhibit more potent efficacy against human lung cancer than ABT-737 but less in platelet reduction in vivo.
- BXI-72 overcomes acquired radioresistance of lung cancer. Based on our findings, the development of BXI(s) as a new class of anticancer agents is warranted and represents a novel strategy for improving lung cancer outcome (Cancer Res. 2013 Sep 1; 73 (17) : 5485–5496) .
- Bax a central death regulator, is required at the decisional stage of apoptosis.
- S184 serine 184
- SMBA1 potently suppresses lung tumour growth via apoptosis by selectively activating Bax in vivo without significant normal tissue toxicity.
- Development of Bax agonists as a new class of anticancer drugs offers a strategy for the treatment of lung cancer and other Bax-expressing malignancies (NATURE COMMUNICATIONS DOI: 10.1038/ncomms5935) .
- BID BH3-interacting domain death agonist
- the level of BID is critical for viability of numerous cells because its silencing makes them resistant to apoptosis induced by death receptor ligands, e.g. TNF-related apoptosis-inducing ligand (TRAIL) .
- TRAIL TNF-related apoptosis-inducing ligand
- tBID appears in cells treated with adenovirus vector expressing the full-length BID (BMC Cancer201414: 771 DOI: 10.1186/1471-2407-14-771) .
- Rapamycin and its derivatives are promising therapeutic agents with both immunosuppressant and anti-tumor properties. These rapamycin actions are mediated through the specific inhibition of the mTOR protein kinase. It is known that mTOR serves as part of an evolutionarily conserved signaling pathway that controls the cell cycle in response to changing nutrient levels.
- the mTOR signaling network contains a number of tumor suppressor genes including PTEN, LKB1, TSC1, and TSC2, and a number of proto-oncogenes including PI3K, Akt, and eIF4E, and mTOR signaling is constitutively activated in many tumor types.
- Rapamycin derivatives have efficacy as anti-tumor agents both alone and when combined with other modes of therapy. Rapamycin appears to inhibit tumor growth by halting tumor cell proliferation, inducing tumor cell apoptosis, and suppressing tumor angiogenesis. Rapamycin immunosuppressant actions result from the inhibition of T and B cell proliferation through the same mechanisms by which rapamycin blocks cancer cell proliferation. Therefore, one might think that rapamycin-induced immunosuppression would be detrimental to the use of rapamycin as an anti-cancer agent.
- rapamycin decreases the frequency of tumor formation that occurs in organ transplant experiments when combined with the widely used immunosuppressant cyclosporine compared with the tumor incidence observed when cyclosporine is used alone.
- the available evidence indicates that with respect to tumor growth, rapamycin anti-cancer activities are dominant over rapamycin immunosuppressant effects.
- clinically rapamycin and its analogs like CCI-779 or temsirolimus, RAD001 or everolimus, Sirolimus, FK-50 and AP23576 have been used in the treatment of various cancers, including kidney cancer, mantle cell lymphoma and metastatic breast cancer.
- Dipyridamole (DPM) like aspirin, inhibits platelet adhesion, and thus tends to prevent the vascular thrombosis of heart attacks and strokes.
- DPM European Stroke Prevention Study
- the introduction to this report reviewed the indicated lack of benefits in treating patients with aspirin who had survived a small stroke, a TIA, a temporary ischemic attack.
- dipyridamole 300 mg a day was added to treatment with aspirin and the results were outstanding. Over a two-year period, stroke deaths were decreased by 50%, deaths from myocardial infarction decreased by 38%and deaths from cancer by 25%.
- dipyridamole may be due only to its prevention of metastases; however, Eva Bestida et al. of the University of Barcelona had a report in Cancer Research, the September 1985 issue (pp. 4, 048-4, 062) on the inhibition of certain human cancer cell growths by dipyridamole. It caused an inhibition of greater than 80%of adinosine, thymidine and uridine. These are substances needed by cancer cells to prosper. This may indicate an anti-cancer effect of dipyridamole other than in the prevention of metastases.
- TRAIL Tumor necrosis factor-related apoptosis-inducing ligand
- RNA interference revealed a preferential dependence on DR5.
- dipyridamole inhibited survivin expression and its important consequences were confirmed by small interfering RNA.
- dipyridamole induced transcriptional shutdown of survivin expression accompanying G1 arrest that was characterized by downregulation of D-type cyclins and cdk6.
- C/EBP CCAAT/enhancer-binding protein
- CHOP homologous protein
- dipyridamole-induced enhancement of TRAIL efficacy and alterations of protein expression were independent of either protein kinase A or protein kinase G.
- findings of the present study describe novel mechanisms of dipyridamole action and highlight its promising use as a potential enhancer of TRAIL efficacy (Oncogene (2008) 27, 3435–3445) .
- DPM Dipyridamole
- Jekunen et al. showed that DPM synergistically enhanced the cytotoxicity of cisplatin in cisplatin-sensitive 2008 human ovarian carcinoma cells by a factor of 4.7, and in the cisplatin-resistant 2008/C13*5.25 subline by a factor of 5.8.
- DPM was found to increase the cellular uptake of cisplatin in a concentration dependent manner, without increasing trypan blue or propidium iodide uptake or changing cell size.
- Gemcitabine (2’, 2’-difluorodeoxycytidine) is a pyrimidine nucleoside analogue. It exerts its cytotoxic effect intracellularly and has activity against a number of different solid tumors, including pancreatic, breast, lung, and bladder cancers. As gemcitabine is strongly hydrophilic, passive diffusion through the hydrophobic cellular plasma membrane lipid bilayer is slow. In order to efficiently enter cells, gemcitabine requires physiologic nucleoside transporter proteins to cross the plasma membrane. These transporter proteins fall into two categories, equilibrative transporters and concentrative transporters.
- hENT human equilibrative nucleoside transporters
- NBMPR nitrobenzylmercaptopurine ribonucleoside
- Fluorouracil (-FU) , sold under the brand name Adrucil among others, is a medication used to treat cancer. By injection into a vein it is used for colon cancer, esophageal cancer, stomach cancer, pancreatic cancer, breast cancer, and cervical cancer. As a cream it is used for actinic keratosis and basal cell carcinoma and as eye drops for treatment of ocular surface squamous neoplasia.
- Dipyridamole has been shown to potentiate the cytotoxicity of 5-FU and other fluoropyrimidines in vitro. This effect probably relates to increases in intracellular concentrations of FdUMP and to the inhibition of the uptake of extracellular nucleosides by the "salvage" pathway. In vitro studies have shown that dipyridamole can augment the cytotoxicity of a number of cytotoxic agents, including etoposide, doxorubicin, vinblastine, and mitoxantrone, perhaps by altering cellular uptake and retention of the cytotoxic agents (Investigational New Drugs 12: 283-287, 1994) .
- Multidrug resistance (MDR) in cancer cells is the simultaneous development of resistance to a variety of anticancer drugs that appear to be structurally and mechanistically unrelated.
- MDR Multidrug resistance
- One type of MDR is characterized by the decreased accumulation of hydrophobic natural product drugs.
- drug efflux is mediated by an adenosine triphosphate (ATP) -dependent membrane transporter termed P-glycoprotein (Pgp) , the product of the MDR1 gene (Juliano and Ling, 1976) .
- Pgp functions as an active outward transport mechanism for a variety of molecules, including certain chemotherapeutic drugs.
- MRP1 The MDR protein 1 (MRP1) gene was cloned from a multidrug-resistant lung cancer cell line and, like Pgp, was found to be a member of the ATP-binding cassette (ABC) superfamily of transporter genes (Cole et al., 1992) . Transfection studies indicated that, similar to MDR1, MRP1 overexpression is sufficient to confer resistance to a broad profile of lipophilic, natural product antineoplastics. MRP1 was the first identified member of a family of genes encoding multispecific organic anion transporter (MOAT) proteins (Borst et al., 1999) .
- MOAT multispecific organic anion transporter
- MRP1 Two other homologues of MRP1, the cMOAT/MRP2 and MRP3 genes, encode proteins that mediate MDR when transfected into a drug-sensitive cell (Borst et al., 1999) . All of these membrane-embedded proteins act as drug efflux pumps, preventing cytotoxic agents from reaching lethal levels within cells.
- P-glycoprotein is a key player in the multidrug-resistant phenotype in cancer.
- the protein confers resistance by mediating the ATP-dependent efflux of an astonishing array of anticancer drugs. Its broad specificity has been the subject of numerous attempts to inhibit the protein and restore the efficacy of anticancer drugs.
- the general strategy has been to develop compounds that either compete with anticancer drugs for transport or act as direct inhibitors of P-gp.
- P-gp-mediated resistance in the clinic. The failure may be attributed to toxicity, adverse drug interaction, and numerous pharmacokinetic issues.
- MRP1 multidrug resistance-associated protein 1
- MRP1 transports a wide range of therapeutic agents as well as diverse physiological substrates and may play a role in the development of drug resistance in several cancers, including those of the lung, breast and prostate, as well as childhood neuroblastoma.
- dipyridamole can significantly increase the cytotoxic and antitumor activities of a variety of chemotherapeutic agents.
- the underlying mechanism here is both prevention of nucleoside and nucleobase salvage, and an increase in the intracellular accumulation of the toxic metabolites via inhibited P-glycoprotein and MRP1 (Clin Pharmacol Ther 2003; 73: 51–60. Drug Metab Dispos. 2014 Apr; 42 (4) : 623–631. Oncogene (2003) 22, 7340–7358) .
- dipyridamole as a single agent in the prevention of tumorigenesis and metastasis in multiple models of triple negative (estrogen and progesterone receptor-negative, Her-2 normal) breast cancers, a subtype that has few effective therapies.
- triple negative (estrogen and progesterone receptor-negative, Her-2 normal) breast cancers a subtype that has few effective therapies.
- These findings provide evidence that intraperitoneal administration of dipyridamole impairs primary tumor growth and metastasis in breast-cancer xenograft animal models.
- our data identify new mechanisms of action of dipyridamole, which is shown to inhibit the ERK1/2-MAPK, NF-kB and Wnt signaling pathways, and to prevent the accumulation of inflammatory cells in the tumor microenvironment.
- dipyridamole was the most potent BCRP inhibitor among the compounds tested with IC50 values of 6.4 +/-0.9 microM. Therefore, dipyridamole has the potential to treat cancer multidrug resistance (Cancer Prev Res (Phila) . 2013 May ; 6 (5) : . doi: 10.1158/1940-6207. CAPR-12-0345) .
- DPM did not improve the anticancer activity of 5-FU or cisplatin in patients with advanced colorectal cancer, metastatic breast cancer, advanced non-small cell lung cancer or advanced measurable pancreatic cancer.
- the observed increase in 5-FU or cisplatin dose-intensity for DPM was not clinically relevant.
- Nanoparticle drug carriers enter cells is different from that of conventional drugs.
- Endocytosis is a process to incorporate extracellular materials into cells. This process can be categorized into three types, i.e., phagocytosis, pinocytosis, and receptor-mediated endocytosis. Phagocytosis only occurs in specialized cells. These cells proliferate and aggregate upon stimulation by extracellular materials and engulf them into lysosomes in the cells for degradation. This process occurs in macrophages and neutrophils of the immune system. Pinocytosis is a process that internalizes extracellular fluid and molecules within it through the invagination of the cell membrane to form a pocket, which then pinches off into the cell to form a vesicle. The vesicle then travels into the cytosol and fuses with other vesicles such as endosomes and lysosomes.
- pinocytosis can be categorized into two types, fluid phase pinocytosis and adsorptive pinocytosis. If the carrier does not have a functional group that interacts with the cells, the cells will engulf the drug carrier by fluid phase pinocytosis. This process is slow and dependent on the carrier concentration around the cell membrane. Adsorptive pinocytosis occurs when the carrier has a hydrophobic group or is positively charged. Such carrier will be physically adsorbed by the cell membrane and increase the engulfing ability of the cells.
- the above two types of endocytosis are non-specific processes and are not suitable for delivery of drugs to their targets. Targeting can only be achieved in certain cancer tissues through enhanced permeability and retention (EPR) .
- EPR enhanced permeability and retention
- Receptor-mediated endocytosis is a process by which cells absorb molecules (endocytosis) by the inward budding of plasma membrane vesicles containing proteins with receptor sites specific to the molecules being absorbed. After the drug carrier binds to the receptor on the cell, an intrinsic signal will trigger the cell membrane to form a coated pit.
- the surface area of a coated pit amounts to 1 to 2%of the cell membrane.
- the coated pit will detach from the cell membrane and enter into the cell to form coated vesicles in the cell, and subsequently form endosomes and move inside the cell in saltatory motion.
- An endosome is a complicated structure comprising microtubules and vesicles. The vesicles can fuse with Golgi. Due to the proton pump (ATPase) , endosomes usually become acidic. The endosomes will then fuse with lysosomes to form secondary lysosomes.
- the cell membrane is a barrier to be overcome for efficient delivery of therapeutics into a target site in mitochondria, cytoplasm or nucleus.
- Hydrophobic phospholipids are major components of the cell membrane that obstruct the transportation of therapeutics.
- various delivery systems such as liposomes, nanoparticles and viral vectors, have been developed to transfer small molecules, peptides, proteins, and oligonucleotides across the membrane. Such manner of drug delivery is herein referred to as cell-penetrating drug delivery systems.
- a number of drug carrier systems have been explored for intracellular delivery of therapeutics. They need to be adapted to cross a series of membrane barriers in order to reach the site of drug action in the cells. During this process, a significant portion of the drug molecules will be lost at each successive barrier. These barriers include cellular association and internalization of the drug-carriers by endocytosis; intracellular trafficking and release of drug or drug-carrier into the cytoplasm; cytoplasmic translocation of drug or drug-carrier to nucleus or any other cellular organelle; and the nuclear/organellar uptake. Cells contain several intracellular organelles with specific functions. Intracellular targeting of therapeutics to these specific organelles is expected not only to significantly enhance the therapeutic efficacy but also reduce non-specific effect and hence toxicity. Therefore, there is significant interest in achieving intracellular target-specific delivery of therapeutics using different carrier systems.
- the carriers that facilitate the endocytosis of drugs include nano-sized polymeric carriers and liposomes.
- nano-sized drug carriers can be categorized into nanoparticles, nanoliposomes, nano suspended particles, solid lipid nanoparticles, magnetic nano-carriers, and the like.
- cell-penetrating peptides CPP
- biodegradable nanoparticles CPP
- viral vectors may also be used as delivery systems for enhancing the penetration of drugs into cells.
- CPPs cell penetrating peptides
- TAT HIV-1 transactivating transcriptional activator
- HSV VP-22 Herpes Simplex virus type-1 transcription factor
- TAT penetration through cell membrane has been shown to be independent of receptors and transporters, and has been suggested to enter the cell by forming an inverted micelle by destabilizing the phospholipid bilayer.
- the main benefit of TAT coupling is that, along with efficient delivery of molecules, biological activity of the coupled molecule is preserved, and the size of the molecule being transported is also not a rate-limiting factor.
- TAT has been suggested not only to enhance intracellular delivery, but also nuclear delivery, and hence has been investigated for nucleic acid delivery.
- TAT peptide conjugated to antisense oligonucleotide has been shown to deliver oligonucleotides to the nucleus.
- TAT peptide After being internalized, TAT peptide has also been found to co-localize inside the Golgi body along with BODIPY-ceramide, which is a marker for Golgi body. Therefore, it is quite possible that there is direct trafficking from the early endosome to the Golgi body without entering the late endosome.
- a secretory pathway could be present where the peptide enters the cytosol from the endoplasmic reticulum.
- Gene therapy has demonstrated significant potential in the treatment of genetic, acquired and neurodegenerative disorders.
- various drug delivery systems and polymers are being investigated such as liposomes, cationic lipid-DNA, polymer complexes.
- TAT peptide conjugation to vectors has been explored.
- Kleeman et al. have demonstrated gene expression in alveolar basal epithelial cells with polyethylenimine (PEI) covalently coupled to TAT through a polyethylene glycol (PEG) spacer, which demonstrated higher transfection efficiencies in vivo in mice lung following intratracheal administration than unconjugated PEG complex.
- PEG polyethylene glycol
- CPPs typically have an amino acid composition that either contains a high relative abundance of positively charged amino acids such as lysine or arginine or has sequences that contain an alternating pattern of polar/charged amino acids and non-polar, hydrophobic amino acids. These two types of structures are referred to as polycationic or amphipathic, respectively.
- a third class of CPPs are the hydrophobic peptides, containing only apolar residues, with low net charge or having hydrophobic amino acid groups that are crucial for cellular uptake.
- the arginine-rich cell-penetrating peptides have been the most widely studied.
- TAT peptide from the HIV transactivator protein TAT, Penetratin, a 16 amino acid domain from the Antennapedia protein of Drosophila, a flock house virus (FHV) coat peptide (sequence 35–49) , and oligoarginines.
- FHV flock house virus
- Biodegradable nanoparticle-mediated intracellular delivery is a dynamic process; involving endocytosis, exocytosis, and sorting into different intracellular compartments. It appears that the NP surface and its interaction with cell surface controls the uptake and intracellular trafficking of biodegradable nanoparticles, and hence that of the encapsulated therapeutic agents.
- Viral vectors are tools commonly used by molecular biologists to deliver genetic materials into cells. This process can be performed inside a living organism (in vivo) or in cell culture (in vitro) . Hence, viral vectors are applicable options for use in cell-penetrating drug delivery systems.
- Cell-penetrating peptides and biodegradable nanoparticles are used not only to modify drugs but also to be conjugated to carries to enhance the transmembrane effects.
- Dipyridamole is an equilibrative nucleoside transporter (ENT) inhibitor.
- Nucleoside transporters (NTs) play an essential role in the transport of nucleosides across cellular membranes. Dipyridamole blocks the equilibrative nucleoside transporter (ENT) , which facilitates the transmembranous diffusion of adenosine. Dipyridamole will increase the extracellular endogenous adenosine concentration, mainly in situations of increased extracellular formation of adenosine, such as occurs during hypoxia or inflammation. However, the extracellular endogenous adenosine concentration induced by dipyridamole causes vasodilatation, which contributes to the metabolic control of organ perfusion.
- Dipyridamole stress myocardial imaging is a widely used and successful technique for diagnosing and evaluating coronary artery disease. Coronary vasodilation with IV dipyridamole is associated with significant reductions in blood flow to collateral-dependent myocardium consistent with coronary steal in patients with CAD. In addition, there have been further studies that discovered vasoconstrictor and vasodilator effects of dipyridamole in many organs, including kidney, lung, pancreas, brain and so on.
- Dipyridamole not only causes vasoconstriction in some organs but can also lead to low blood pressure and subsequent side effects such as vertigo and palpitations due to dilation of blood vessels of the heart.
- the effect of reducing blood pressure makes dipyridamole unsuitable for the treatment of patients who are physiologically unstable, such as those having, but not limited to, sepsis, ischemic stroke, hemorrhagic stroke, acute lung injury, acute liver injury, myocardium infarct, and cardiorenal syndrome.
- the blood-flow restricting effect of dipyridamole limits its application in the treatment of diseases involving organs rich with blood vessels.
- dipyridamole Since the pharmacological action of dipyridamole is mainly on cell membranes, a delivery system designed for membrane penetration avoiding binding with equilibrative nucleoside transporter on the membrane while enhancing the intracellular signal transduction and PPAR ⁇ regulation can prevent the effect of tissue hypoperfusion due to increased cardiovascular dilation and local blood flow restriction. The limitation in clinical applications of dipyridamole in acute and severe patients due to the decrease of blood pressure can thus be lifted.
- Dipyridamole is also a non-selective phosphodiesterase inhibitor. Increase of intracellular drug delivery will enhance the inhibition of dipyridamole on intracellular phosphodiesterase (PDE) .
- PDE intracellular phosphodiesterase
- Members of the PDE family have unique cell-and tissue-specific distribution. Dipyridamole may be used as anti-inflammatory, anti-oxidant, anti-fibrosis, and smooth muscle relaxing agents for treating diseases associated with PDE regulation depending on the distribution profile of PDE on cell membranes or in cytoplasm in different tissues.
- dipyridamole may be used for treating lower urinary tract dysfunction and erectile dysfunction, like other PDE5 inhibitors.
- dipyridamole since dipyridamole is a non-selective PDE inhibitor, it may be used for the treatment of PDE associated diseases when delivered by a transmembrane drug delivery system.
- CD437 is able to induce apoptosis in a variety of human carcinoma cells in vitro and in vivo. In intact cells, CD437-dependent caspase activation is preceded by the release of cytochrome C from mitochondria. Moreover, when added to isolated mitochondria, CD437 causes membrane permeabilization.
- CD437 represents a low molecular weight compound which exerts its cytotoxic effect via the mPTPC, i.e., by acting directly at the surface or inside of mitochondria.
- arsenic trioxide which is used in the treatment of acute promyelocytic leukemia, has multiple actions on mitochondria. It is known to cause the induction of mPTPC formation via its action on the voltage-dependent anion channel VDAC. Arsenic trioxide is also known to act on the respiratory chain and inhibit respiratory chain activity.
- Apoptotic factors that play a major role in the modulation of apoptosis include Bcl-2 and Bcl-Xl. Compounds that act by binding to these proteins have been identified and studied for their efficacy; a few examples include a chromene derivative and gossypol which was recently shown to act on proteins of the Bcl-2 family. In fact there are so many varied and structurally different compounds that it has been suggested that they be collectively called mitocans to reflect their mitochondrially mediated anticancer effects.
- the selective accumulation approach to targeting tumor mitochondria requires two levels of specific accumulation; drug accumulation in the tumor and then drug accumulation in the mitochondria of cancer cells.
- drug disposition may be modulated by subtle modification of the chemical structure to change its physico-chemical properties that are known to determine its accumulation in various compartments. Of course such modification must be done without adversely affecting action on the molecular target.
- the second approach involves conjugating ligands that are larger than simple organic functional groups to change the biodistribution of the active molecule. Again this approach works as long as the conjugation does not adversely affect the desired pharmacological activity of the molecule.
- Such approaches have been used very effectively to alter drug distribution in the body and achieve higher accumulation in target tissues using ligands that are known to have an affinity for the target tissue.
- nanocarriers like liposomes, micelles, and solid nanoparticles offer what might be viewed as a non-chemical approach to modify the disposition of drug molecules. All chemistry can be performed on the components of the nanocarrier system that can then be loaded with the drug to afford targeted delivery. Most pharmaceutical nanocarriers can be modified for targeting to specific tissues and even specific cell types. Long-circulating liposomes and nanoparticles are able to passively target areas of leaky vasculature by virtue of the enhanced permeability and retention (EPR) effect and can additionally be modified with antibodies or other targeting ligands to afford cell-specific recognition.
- EPR enhanced permeability and retention
- Nanocarriers that can not only affect the tumor-specific accumulation of a drug but also mediate mitochondria-specific accumulation within a tumor cell might be the ultimate tool in mitochondria-targeted anticancer approaches if they can be developed for clinical therapy. The first steps in this direction have already been taken in recent years. Current nanocarrier technology is reaching the point where the need for sub-cellular delivery may indeed be met using nanocarriers specifically designed for such purposes.
- liposomes were modified with both transferrin and a fusogenic peptide Chol-GALA.
- the transferrin modification enhances liposomal uptake into cells via endocytosis after which the peptide facilitates release from the endosomes into the cytosol.
- the delivery approach achieved a higher concentration of the drug that was potentially available to interact with the sub-cellular target.
- Micelles have also been proposed for the delivery of hydrophobic drugs to various sub-cellular organelles including mitochondria.
- the fluorescently labeled micelles used in the study were found to be distributed through several cytoplasmic organelles, including a majority of them associated with the mitochondria. The uptake of these micelles was not restricted to a single cell type. Also, the extent of cell internalized cargo incorporated in micelles was greater than the free cargo by itself.
- nanocarriers designed specifically to accumulate in mitochondria. Arguably the earliest of these are what are known as DQAsomes. Prepared from the mitochondriotropic molecule dequalinium chloride, these vesicular nanocarriers were developed for mitochondria-specific DNA delivery but were also shown to be capable of changing the sub-cellular distribution of paclitaxel to increase the accumulation of the drug in mitochondria. The mitochondria-specific delivery led to improved apoptotic activity at paclitaxel concentrations, at which the free drug does not have a significant cytotoxic effect.
- Paclitaxel loaded DQAsomes have also been tested for their ability to inhibit the growth of human colon cancer tumors in nude mice and the data strongly suggest that encapsulation of paclitaxel in DQAsomes leads to improved efficacy.
- the antitumor efficiency of DQAsomal encapsulated paclitaxel was also further enhanced by modifying the DQAsomal surface with folic acid (FA) .
- the folate receptor is a folate high-affinity membrane binding protein, which is overexpressed in a large variety of human tumors.
- FA conjugates are internalized in a tumor cell-specific manner by receptor-mediated endocytosis resulting in an increased toxicity of the corresponding drug.
- TPP again served as the mitochondriotropic ligand in liposomal and polymer based nanocarriers.
- Liposomes have been well characterized as delivery systems and are a popular choice due to their biocompatibility, ease of surface modification, and capacity to encapsulate hydrophilic or hydrophobic drugs.
- the first indication that liposomes could be rendered mitochondriotropic by surface modification with a mitochondriotropic residue came from a report on so-called proteoliposomes prepared by incorporating a crude mitochondrial membrane fraction into liposomes colocalized with endogenous mitochondria in pre-implantation embryos. Further investigation of the concept of using ligands to alter the sub-cellular distribution of liposomes was based on the synthesis of stearyltriphenylphosphonium (STPP) .
- STPP stearyltriphenylphosphonium
- STPP-liposomes were shown to effectively direct the accumulation of rhodamine labeled phosphatidylethanolamine (Rh-PE) to mitochondria in live cells. Based on flow cytometry, STPP-liposomes exhibited the same level of cell association as liposomes with the same cationic charge. However, the subsequent sub-cellular localization analyzed by confocal microscopy was markedly different, indicating that the mitochondriotropic ligand, and not the surface charge, is what determines mitochondria-specific association of the nanocarrier. It was also found that the TPP ligand did not change the in vivo distribution and tumor accumulation of long-circulating PEGylated liposomes.
- STPP-liposomes did, however, improve the both the in vitro and the in vivo efficacy of ceramide. Taken together these data suggest that the tendency of long-circulating liposomes to passively accumulate (via the EPR effect) in solid tumors can be combined with organelle-specific tropism conferred by modification with an appropriate ligand to potentiate the effect of an encapsulated antitumor agent.
- MITO-Porter an alternative approach to developing mitochondria-specific liposomes has focused on the concept that liposomes with a tendency to selectively fuse with mitochondrial membranes are more likely to associate with mitochondria upon cell entry.
- these liposomes are surface modified with octaarginine residues to facilitate their entry into cells as intact vesicles (via macropinocytosis) .
- the lipid composition was selected on the basis of high levels of fusion with the mitochondrial membrane and the release of its cargo to the intra-mitochondrial compartment in living cells. Based on confocal microscopy data, MITO-porter liposomes have been used to deliver green fluorescent protein as well as propidium iodide to mitochondria, suggesting that they can be used to deliver macromolecules as well as small molecules to mitochondria.
- mitochondria-specific nanocarriers has not been limited to lipid based carriers but also includes the use of mitochondriotropic residues to create polymeric systems capable of mitochondria-specific intracellular delivery of bioactive molecules.
- TPP modification has been employed to create a mitochondriotropic N- (2-hydroxylpropyl) methacrylamide (HPMA) copolymer-based nanoparticle.
- HPMA mitochondriotropic N- (2-hydroxylpropyl) methacrylamide
- HPMA copolymer-drug conjugates were synthesized using a photosensitizer mesochlorin e 6 (Mce 6) .
- Mce 6 photosensitizer mesochlorin e 6
- Mitochondrial targeting of HPMA copolymer-bound Mce 6 enhanced cytotoxicity as compared to non-targeted HPMA copolymer- Mce 6 conjugates.
- the authors indicate that “minor modifications may be required to adapt the current design and allow for tumor site-specific mitochondrial targeting of other therapeutic agents. ” Therefore these systems could theoretically be applied to the mitochondria-specific delivery of a range of pro-apoptotic substances for cancer therapy.
- Inorganic nanoparticles have also been shown to be capable of mitochondria-specific delivery.
- hypocrellin A a photodynamic drug
- HANC water-soluble amorphous silica nanocage
- These drug-loaded nanocages are reportedly able to specifically accumulate in the mitochondria of cancer cells and improve the photosensitizing effect of hypocrelin A. It is, however, unclear what mediates the mitochondria-specific accumulation of the nanocage system. Nevertheless, taken together, the various studies described so far strongly support the hypothesis that nanocarriers can indeed control the sub-cellular accumulation of bioactive molecules and as such represent a useful tool in the development of mitochondria-targeted anticancer strategies (G. G. M. D'S ouza et al. /Biochimica et Biophysica Acta 1807 (2011) 689–696) .
- Adenosine downregulated the expression of mRNAs and proteins for Bcl-XL and inhibitor of apoptosis protein 2 (IAP2) to directly inhibit caspase-3, -7, and -9, but it otherwise upregulated the expression of mRNA and protein for DIABLO, an inhibitor of IAPs.
- IAP2 apoptosis protein 2
- Extracellular adenosine induces apoptosis in a variety of cancer cells via intrinsic and extrinsic pathways.
- adenosine uptake into cells triggers apoptosis
- adenosine receptors mediate apoptosis. Dipyridamole inhibiting the cellular uptake of adenosine reversed significantly the adenosine-induced growth suppression.
- the present invention provides a novel type of BAX/BCL-2 modulator having the pyrimido [5, 4-d] pyrimidine main structure and a method of preventing or treating BAX/BCL-2-related disorders or conditions, such as cancers, myeloproliferative disorder, prostate epithelial dysplasia, lymphangioleiomyomatosis, Kimura disease, and keloid, using such BAX/BCL-2 modulators.
- the invention also relates to a method of increasing the expression and activity of BAX/BCL-2.
- dipyridamole exhibits anticancer activity, the efficacy is insignificant at low dose.
- Dipyridamole is a known ENT-1 inhibitor which blocks the uptake of adenosine into the cells. Since adenosine may promote apoptosis of cancer cells, such blockade decreases the anticancer efficacy of dipyridamole.
- dipyridamole may be encapsulated in a carrier to facilitate penetration of the cell membrane and accumulation in the cells, and thereby activate the endogenous apoptosis mechanism of the cells.
- dipyridamole in the cells may be achieved through increasing the formation of BAX/BCL-2 heterodimer, increasing the formation of BAX homodimer and decreasing the formation of BCL-2 homodimer. It is also found in the present invention that dipyridamole can reduce BCL-XL expression, reduce the inhibition of ENT-1 and modulate the apoptotic protein expression of the cancer cell, thereby increasing its anticancer ability.
- dipyridamole is encapsulated in a cell-penetrating carrier to increase the concentration of dipyridamole inside the cell and reduce the inhibition on ENT-1.
- increase of BCL-XL and decrease of cytosolic adenosine, and thus decrease of anticancer efficacy can be avoided.
- the invention relates to a method of preventing or treating a BAX/BCL-2-related disorder or condition, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of formula (I) :
- each of R 1 , R 2 , R 3 and R 4 is independently selected from the group consisting of heterocyclyl and di (hydroxyalkyl) amino,
- the present invention also relates to use of a compound of formula I or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for preventing or treating BAX/BCL-2-related disorders or conditions.
- the medicament comprises a compound of formula I or a pharmaceutically acceptable salt thereof encapsulated in a pharmaceutically acceptable carrier.
- the present invention further relates to a pharmaceutical composition for preventing or treating BAX/BCL-2-related diseases, comprising a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt thereof encapsulated in a pharmaceutically acceptable carrier.
- a pharmaceutical composition for preventing or treating BAX/BCL-2-related diseases comprising a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt thereof encapsulated in a pharmaceutically acceptable carrier.
- the compound is dipyridamole and the carrier is a liposome.
- Figures 1a and 1b are schemes showing the different actions of dipyridamole (a) outside the cell and (b) inside the cell on cancer cell apoptosis.
- Figure 2 demonstrates the correlation between cell viability and Bax/Bcl-2 ratio (Y-axis: cell viability; X-axis: Bax/Bcl-2 ratio) .
- FIG. 3 shows the structures of representative Rapalogs.
- Figures 4a and 4b show the cell viability of the triple-negative breast cancer cell line MDA-MB-231 treated by dipyridamole with/without 5-FU.
- FIGS 5a and 5b show the Bax, Bcl-2 and Bcl-xL expression of cancer cells treated by dipyridamole with/without 5-FU.
- FIGS 6a and 6b show the Bax/Bcl-2 ratio in cells treated by dipyridamole with/without 5-FU.
- BAX/BCL-2 modulators refers to the agents that can modulate the expression or activity of BAX/BCL-2 homodimer.
- alkyl refers to a saturated straight-chain or branched hydrocarbon group having 1 to 6 carbon atoms, especially 1 to 4 carbon groups, for example methyl, ethyl, propyl, 1-methylethyl, butyl, 1-methylpropyl, 2-methylpropyl, 1, 1-dimethylethyl, pentyl, 1-methylbutyl, 2-methylbutyl, 3-methylbutyl, 2, 2-dimethylpropyl, 1-ethylpropyl, hexyl, 1, 1-dimethylpropyl, 1, 2-dimethylpropyl, 1-methylpentyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 1, 1-dimethylbutyl, 1, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2, 2-dimethylbutyl, 2, 3-dimethylbutyl, 3, 3-dimethylbutyl, 1-ethylbutyl, 2-ethylbutyl
- heterocyclyl refers to a monocyclic radical having 5 to 8 ring members, wherein in each case 1, 2, 3 or 4 of these ring members are heteroatoms selected, independently from each other, from the group consisting of oxygen, nitrogen and sulfur.
- preventing refers to delaying the onset of the symptoms of a susceptible subject or reducing the occurrence of a disease.
- treating denotes reducing and/or improving the symptoms of a susceptible subject or increasing the survival rate of the subject with certain lethal disorders or conditions.
- BAX/BCL-2-related disorders or conditions denotes the disorders or conditions wherein the modulation of BAX/BCL-2 is beneficial.
- disorders or conditions include cancers, myeloproliferative disorder, prostate epithelial dysplasia, lymphangioleiomyomatosis, Kimura disease, and keloid.
- subject denotes animals, especially mammals. In one preferred embodiment, the term “subject” denotes humans. In another preferred embodiment, the term “subject” denotes companion animals, such as cats and dogs.
- terapéuticaally effective amount refers to the amount of an active ingredient used alone or in combination with other treatments/medicaments for treating PPAR ⁇ -related disorders or conditions that shows therapeutic efficacy.
- carrier or “pharmaceutically acceptable carrier” refers to particles that can encapsulate active pharmaceutical ingredients.
- carriers suitable for the present invention include niosomes, polymersomes, nanoparticles, liposomes, nano suspended particles, solid lipid nanoparticles, magnetic nano-carriers, micelles, macromolecular conjugates, particulate drug carriers, and the like.
- the inventors of the invention surprisingly found that compounds having a pyrimido [5, 4-d] pyrimidine structure can enhance the expression and activity of BAX/BCL-2 homodimer, and thus may serve as novel types of BAX/BCL-2 modulators.
- the pyrimido [5, 4-d] pyrimidine compound is dipyridamole.
- the present invention thus provides a method of preventing or treating BAX/BCL-2-related disorders or conditions, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of formula (I) :
- each of R 1 , R 2 , R 3 and R 4 is independently selected from the group consisting of heterocyclyl and di (hydroxyalkyl) amino,
- R 1 and R 3 are heterocyclyl, preferably piperidyl, and R 2 and R 4 are di(hydroxyalkyl) amino, preferably N, N-di (hydroxyethyl) amino.
- the compound is dipyridamole.
- the compound is encapsulated in a carrier, such as a niosome, a polymersome, a nanoparticle, a liposome, a nano suspended particle, a solid lipid nanoparticle, a magnetic nano-carrier, a micelle, a macromolecular conjugate or a particulate drug carrier.
- a carrier such as a niosome, a polymersome, a nanoparticle, a liposome, a nano suspended particle, a solid lipid nanoparticle, a magnetic nano-carrier, a micelle, a macromolecular conjugate or a particulate drug carrier.
- the carrier is a liposome.
- the liposome has a diameter in the range of about 50-700 nm, preferably about 60-530, more preferably 80-350 nm, and most preferably about 130-230 nm.
- dipyridamole when dipyridamole is administered in free form, it binds to the ENT receptors on a cell membrane and activates signaling pathways that block adenosine from passing into the cell.
- dipyridamole can regulate the expression of apoptosis/anti-apoptosis proteins and cause cell apoptosis through cell-penetrating drug delivery systems into the cell. Activation of the apoptosis pathway of cancer cells can facilitate the treatment of cancers known to be associated with abnormal apoptosis/anti-apoptosis protein expression.
- Figure 1a shows that if dipyridamole is administered in free form, it mainly acts outside of cells and will promote the accumulation of adenosine, which will lead to reduced apoptosis.
- Figure 1b shows that dipyridamole inside the cell can activate the expression of apoptosis/anti-apoptosis proteins. In such case, the unfavorable action of dipyridamole outside the cell can be avoided.
- the present invention demonstrates that by delivering dipyridamole directly into cells, the binding to the receptors on the cell membrane can be avoided so as to reduce the side effects such as oxidative stress and vasoconstriction caused by the accumulation of adenosine.
- the compound of formula (I) of the invention is encapsulated in a carrier for delivery into the cell.
- the carrier is a niosome, a polymersome, a nanoparticle, a liposome, a nano suspended particle, a solid lipid nanoparticle, a magnetic nano-carrier, a micelle, a macromolecular conjugate or a particulate drug carrier.
- the carrier is a liposome.
- the liposome suitable for the present invention has a diameter in the range of about 10-300 nm, preferably about 80-280 nm, more preferably about 120-270 nm.
- the carriers may be niosomes, polymersomes, or polymers that have a diameter of less than 1 ⁇ m. Modifications can be made based on surface electric potential, hydrophilicity/hydrophobicity, size, morphology, shape and/or surface curvature.
- the liposome formulation of the invention may comprise vesicles of various natures (e.g., unilamellar or multilamellar) , compositions, sizes, and characteristics, enclosing an aqueous medium of diverse compositions, pH and osmotic strength.
- the main constituents of the liposome lipid layer membrane are selected from the group consisting of natural or synthetic phospholipids such as those listed below:
- DOPC 2-Dioleoyl-sn-Glycero-3-Phosphocholine
- DOPE 2-Dioleoyl-sn-Glycero-3-Phosphoelhanolamine
- MPPC -1-Myristoyl-2-Palmitoyl-sn-Glycero-3-Phosphocholine
- SPPC -1-Stearoyl-2-Palmitoyl-sn-Glycero-3-Phosphocholine
- Preferred phospholipids are long saturated phospholipids, e.g. those having alkyl chains of more than 12, preferably more than 14, more preferably more than 16, most preferably more than 18 carbon atoms.
- Preferred liposome compositions for use according to the invention are preferably those in which the liposomes are uni-and/or multilamellar, and comprise:
- phospholipids 1 to 100, preferably 40 to 70 mol%physiologically acceptable phospholipids, preferably selected from the group consisting of DLPC, DMPC, DPPC, DSPC, DOPC, DMPE, DPPE, DSPE, DOPE, MPPC, PMPC, SPPC, PSPC, DMPG, DPPG, DSPG, DOPG, DMPA, DPPA, DPPS, PS, EPC, SPC and HSPC;
- (iii) 1 to 100, preferably 40 to 70 mol%surfactants, preferably featuring hydrophobic alkyl ethers (e.g. Brij) , alkyl esters, polysorbates, sorbitan esters, and/or alkyl amides;
- hydrophobic alkyl ethers e.g. Brij
- alkyl esters e.g. polysorbates, sorbitan esters, and/or alkyl amides
- liposome-like vesicles are made from polymers and comprise no lipids, for which reason they are formally not considered liposomes but are called polymersomes.
- polymersomes are meant to be encompassed by the term liposome as used for defining the invention and the claims.
- niosomes liposome-like vesicles made from synthetic surfactants and comprising no lipids are called niosomes.
- niosomes are meant to be encompassed by the term liposome as used for defining the invention and the claims.
- polymerization of different high molecular polymers can be used, which comprise those in tri-block copolymer form such as ABA and BAB, and those in block copolymer form such as PLLA-PEG, PLGA-PEG, PLA-PEG, PLLA-mPEG, PLGA-mPEG and PLA-mPEG.
- Various shapes such as asterisk and L form can be designed, including block copolymers of PEG- (PLGA) 8 , PEG- (PLLA) 8 and PEG- (PDLA) 8 Star.
- PEGylated modification can be used to modify any vehicle such as polymeric vehicle and liposome to achieve the effect of reducing the binding rate of plasma proteins (see Park, J.
- the animal dose should not be extrapolated to a human equivalent dose (HED) by a simple conversion based on body weight.
- HED human equivalent dose
- the Food and Drug Administration has suggested that the extrapolation of animal dose to human dose is correctly performed only through normalization to BSA, which often is represented in mg/m2.
- the effective dose of liposome-dipyridamole in mice is 10 mg/kg-100 mg/kg, in hamster 6-60 mg/kg, in rat 5-50 mg/kg, in guinea pig 3.75-37.5 mg/kg, in rabbit 2.5-25 mg/kg, in monkey 2.5-25 mg/kg, in dog 1.5-15 mg/kg, in cat 2.4-24 mg/kg, in baboon 1.5-15 mg/kg, in child 1.2-12 mg/kg, and in adult 0.81-8.1 mg/kg.
- the broadest dose range without limiting the species is 0.4-160 mg/kg, preferably 0.6-120 mg/kg, more preferably 0.8 mg/kg-100 mg/kg.
- the dipyridamole liposome may be used for treating cancer.
- the cancers are drug-resistant or non-drug-resistant.
- the cancers are breast cancers and liver cancers.
- the dipyridamole liposome may be administered in combination with other anti-cancer drugs, such as alkylating agents, antimetabolites, antibiotics, hormones, immune modulators, mitotic inhibitors, target therapeutic agents, and platinum drugs.
- the anti-cancer drugs comprise chemotherapeutic drugs and target drugs.
- the chemotherapeutic drugs comprise alkylating agents (such as Cyclophosphamide, Mechlorethamine and Melphalan ) , antimitotic agents (Vinblastine, Vincristine and Taxol) , DNA intercalating agents (Daunorubicin and Doxorubicin) , DNA cleaving agents, antimetabolites agents (such as Capecitabine, Cladribine, Cytarabine, Fludarabine phosphate, 5-Fluorouracil, Gemcitabine, 6-Mercaptopurine, Methotrexate (Amethopterin,MTX) , Mitoxantron, Pemetrexed disodium (Heptahydrate) , and Tegafur (FT-207) ) and topoisomerase inhibitors (such as (Camptothecin, Topotecan, Irinotecan, Podophyllotoxin and Etoposide) .
- alkylating agents such as Cyclophosphamide, Mechloreth
- the target drugs comprise Trastuzumab, Lapatinib, Genfitinib, Erlotinib, Cetuximab, Bevacizumab, Rituximab, Bortizomib, Imatinib, Sunitinib, Rapamycin, Sirolimus, Temsirolimus, Everolimus, Ridaforolimus (deforolimus) and Sorafenib.
- the other anti-cancer drug is 5-Fluorouracil (5-FU) .
- Liposomes were prepared with positive and neutral charge containing phospholipid and cholesterol, wherein the mole percent of cholesterol was 5%to 75%. Small unilamellar vesicles were prepared. The dried lipid films were hydrated with an ammonium sulfate and sequentially extruded through a series of polycarbonate membrane filters. Dipyridamole was encapsulated into the liposomes via a transmembrane pH gradient or dehydration-rehydration, and the diameters of the extruded liposomes were in the range of 50-602 nm (dissolved in glucose solution) via the above manufacturing processes. The diameter of the liposome-dipyridamole was about 50 to 602 nm as shown in Table 1.
- Example 2 Cell viability (MTT assay) and Western blot analysis of the BCL-2 protein family
- the medium for human breast adenocarcinoma is L-15 Medium. To make the complete growth medium, the following components were added to the base medium: fetal bovine serum to a final concentration of 10%. The cultures were incubated at 37°C without CO 2 .
- the tested cancer cell lines include MDA-MB-231, PANC-1, BXPC-3, HCT-116, HT29, A375, and MeWo.
- MTT (3- (4, 5-dimethylthiazol-2-yl) -2, 5-diphenyl-tetrazolium bromide, Sigma, St. Louis) was prepared by dissolving 5 mg MTT/ml in phosphate-buffered saline pH 7.5 and filtered through a 0.22 ⁇ m filter.
- the MTT assay used was essentially similar to that originally described by Mosmann (1983) . Briefly, 100 ⁇ L of MTT solution (0.5 mg/mL) was added to the monolayers of the treated cancer cell lines and the microplates were incubated at 37°C for 3h. 100 ⁇ L of DMSO was added to each well and mixed thoroughly to dissolve the dark blue crystals. The plates were read on a microELISA reader using a test wavelength of 570 nm and reference wavelength of 630 nm.
- the Western blot method was performed as follows: protein concentration was measured using Bradford assay; 6X sample buffer (0.8 mM Tris-HCl, 10 mM EDTA, 10%SDS, 60%glycerol, 0.6 M ⁇ -mercaptoethano, 0.06%bromophenol blue, pH 6.8) was added into 50 ⁇ g of whole cell proteins; and an equal volume of lysis buffer was added to the samples. After being heated at 95°C for 10 minutes to denature the proteins, the samples were immediately cooled on ice.
- 6X sample buffer 0.8 mM Tris-HCl, 10 mM EDTA, 10%SDS, 60%glycerol, 0.6 M ⁇ -mercaptoethano, 0.06%bromophenol blue, pH 6.8
- the samples were then separated by 12%SDS-PAGE electrophoresis (100 V) and transferred from the SDS-PAGE gels to PVDF membranes by wet blotting.
- the PVDF membranes were then treated with 5%skimmed milk at room temperature for 60 minutes to block non-specific binding.
- the membranes were incubated with primary antibody overnight at 4°C and washed three times with PBST.
- the membranes were incubated with a secondary antibody at room temperature for 60 minutes and washed three times with PBST.
- the membranes were then washed one more time with PBS and incubated with an enhanced chemiluminescence (ECL) substrate for detection.
- Photos of the images were taken using automated chemiluminescence and fluorescence imaging system (UVP Biospectrum) .
- Example 2.1 Cell viability and Bax/BCL-2 ratio of MCF-7 in cells treated with the dipyridamole liposome
- the cell line used in the assay was human breast adenocarcinoma cells, MCF-7.
- the cells were treated with the dipyridamole liposome (10 and 20 ⁇ M) or dipyridamole freeform (10 and 20 ⁇ M) .
- the results are shown in Figures 4a and 4b, and normalized in Figures 5a and 5b and Table 2.
- Example 3 Cell viability and Bax/BCL-2 ratio of MDA-MB-231 in cells treated with the dipyridamole liposome in combination with Rapamycin
- the cell line used in the assay was human breast adenocarcinoma cells, MDA-MB-231.
- the cells were treated with rapamycin (100 ⁇ M) , the dipyridamole liposome (3.125 and 25 ⁇ M) or dipyridamole freeform (3.125 and 25 ⁇ M) in combination with rapamycin (100 ⁇ M) .
- the results are shown in Table 3.
- Example 4 Cell viability and Bax/BCL-2 ratio in PANC-1 cells treated with the dipyridamole liposome in combination with Rapamycin
- the cell line used in the assay was human pancreas adenocarcinoma cells, PANC-1.
- the cells were treated with rapamycin (10 ⁇ M) , the dipyridamole liposome (3.125 and 25 ⁇ M) or dipyridamole freeform (3.125 and 25 ⁇ M) in combination with rapamycin (10 ⁇ M) .
- the results are shown in Table 4.
- Example 5 Cell viability and Bax/BCL-2 ratio in PANC-1, BxPC-3, HCT-116, HT29, A375 and MeWo cells treated with the dipyridamole liposome in combination with 5-Fu
- the cells were treated with 5-Fu (10 ⁇ M and 100 ⁇ M) , the dipyridamole liposome (3.125 and 25 ⁇ M) or dipyridamole freeform (25 ⁇ M) in combination with 5-Fu (10 ⁇ M) .
- the results are shown in Table 5.
- results in this example demonstrate that by using dipyridamole liposome in combination with 5-Fu, the cytotoxic effect of these drugs on pancreatic, liver, breast and skin cancer cells is increased through balancing Bcl-2 and Bax ratio.
- the doses of these drugs may also be reduced.
- the dose of 5-Fu may be reduced by at least 8 fold.
- Example 6 Cell viability of A375 and MeWo cells treated with the dipyridamole liposome in combination with Gemcitabine
- dipyridamole liposome exhibits a membrane-penetrating effect.
- dipyridamole reduces antagonism to Gemcitabine as effective by 20-100%.
- This result proves that dipyridamole liposome increases penetration of drugs into the cells.
- Table 6 Extracellular dipyridamole blocks ENT-1 and prevents Gemcitabine from entering into the cancer cells to produce its cytotoxic effect.
- the results in this example demonstrate that dipyridamole liposome reduces this phenomenon by 20-40%, indicating that dipyridamole liposome enhances the ratio of drugs that enter the cells.
- chemotherapeutic drugs for example, the rapamycin analogs (Rapalogs) shown in Figure 3, may be used in combination with dipyridamole liposome of the present invention.
- dipyridamole liposome of the present invention produces a synergistic effect in inhibiting cancer cell proliferation when administered together with a chemotherapeutic drug by 10-40%more cell death (Fig. 4) .
- the synergistic effect in cancer cell death may result not simply from increasing Bax or decreasing Bcl-2, but increasing the Bax/Bcl-2 ratio (Figs. 5 and 6) .
- dipyridamole increases Bax/Bcl-2 ratio by 25-200%after entering into the cells, and increases inhibition on cancer cell proliferation by 20-50%.
- dipyridamole inhibits cancer cell proliferation by 5-50%through entering into the cells and increasing Bax/Bcl-2 ratio by 15-50%.
- Combination of dipyridamole with rapamycin inhibits cancer cell proliferation by 10-30%through entering into the cells and increasing Bax/Bcl-2 ratio by 10-200%.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Veterinary Medicine (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Epidemiology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Organic Chemistry (AREA)
- Molecular Biology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Medicinal Preparation (AREA)
Abstract
The use of a therapeutically effective amount of a compound of formula (I) in preparing medicines for preventing or treating BAX/BCL-2-related disorders, including cancers, myeloproliferative disorder, prostate epithelial dysplasia, lymphangioleiomyomatosis, Kimura disease and keloid.
Description
The invention provides a method of enhancing the expression and activity of BAX/BCL-2 and related therapeutic use.
The BCL-2 protein family determines the commitment of cells to apoptosis, an ancient cell suicide programme that is essential for development, tissue homeostasis and immunity. These proteins govern mitochondrial outer membrane permeabilization (MOMP) and can be either pro-apoptotic (Bax, BAD, Bak and Bok among others) or anti-apoptotic (including Bcl-2 proper, Bcl-xL, and Bcl-w, among an assortment of others) . There are a total of 25 genes in the Bcl-2 family known to date. Human BCL-2 was discovered as the gene located near the junction at which chromosomes 18 and 14 (t14; 18) are joined anomalously in the tumor cells of follicular lymphoma patients. This chromosome translocation leads to misregulation of the normal BCL-2 expression pattern to contribute to cancer (Tsujimoto et al. 1985; Nunez et al. 1989) .
A variety of physiological death signals, as well as pathological cellular insults, trigger the genetically programmed pathway of apoptosis (Vaux and Korsmeyer 1999) . Apoptosis manifests in two major execution programs downstream of the death signal: the caspase pathway and organelle dysfunction, of which mitochondrial dysfunction is the best characterized (for reviews, see Green and Reed 1998; Thornberry and Lazebnik 1998) . As the BCL-2 family members reside upstream of irreversible cellular damage and focus much of their efforts at the level of mitochondria, they play a pivotal role in deciding whether a cell will live or die.
Recent studies of mitochondrial involvement in cancer have uncovered a plethora of differences in the structure and function of these organelles upon comparing metastatic mitochondria to those belonging to nontransformed cells. Notably, modern research has largely upheld the metabolic observations of Warburg and his successors, while refining and greatly expanding the breadth of mechanistic knowledge of mitochondrial state and function in tumour
development. The comprehensive mechanisms of the Warburg effect have not yet been isolated; however, multiple intertwining causative and responsive mechanisms have recently been characterized. This understanding of the indicative features of cancer cell metabolism has also been directly applied to current clinical care through the increasingly widespread adoption of positron emission tomography (PET) imaging using glucose analogues to identify cancerous lesions that are characterized by high glucose uptake (BioMed Research International Volume 2013 (2013) , Article ID 612369) .
Apoptosis can be initiated via two pathways, the extrinsic and the intrinsic pathway. Whereas the extrinsic pathway is initiated through stimulation of cell surface death receptors, the intrinsic pathway is activated by many different intracellular stresses and is elicited by most chemotherapy agents. The BCL-2 family proteins are predominantly involved in the intrinsic pathway, in which they regulate mitochondrial outer membrane permeabilization (MOMP) . MOMP results in the release of apoptosis-triggering factors, such as cytochrome c, from the mitochondrial intermembrane space into the cytoplasm in which they activate a cascade of caspases that execute widespread proteolytic events leading to cellular demise (3) . MOMP marks commitment to death and is often considered to be the point of no return for a cell. Here, we review the rationale and preliminary results of recent efforts to elicit MOMP by exploiting BCL-2 family proteins as an anticancer therapy and discuss other related targets for drug development (Clin Cancer Res; 21 (12) June 15, 2015) .
Overexpression of Bcl-2 was subsequently described in prostate, breast and colon carcinomas, as well as glioblastomas. Overexpression of Mcl-1, another antiapoptotic, Bcl-2-related protein, was identified in relapsed acute myelogenous leukemia, and was associated with poor prognosis. Other changes in Bcl-2-related protein expression identified in cancer cells include different mutations in the Bax gene, and changes in the proapoptotic to antiapoptotic Bcl-2 protein ratio (Bettaieb et al., 2003; and Oncogene (2004) 23, 2934–2949) .
Overexpression of the Bcl-2 and related antiapoptotic proteins has been demonstrated to inhibit cell death induced by many stimuli, including growth factor deprivation, hypoxia and oxidative stress. However, it is the ability of antiapoptotic Bcl-2-family proteins to suppress cell death induced by cytotoxic anticancer drugs that makes these proteins particularly interesting as potential targets for cancer drug discovery. Regardless of the primary mode of action, whether single or double-strand DNA breaks, whether microtubule depolymerization or aggregation,
whether nuclear hormone receptor activation (glucocorticoid receptor) or inhibition (estrogen and androgen receptors) , essentially all traditional anticancer drugs appear to depend in large measure on Bcl-2/Bax-dependent mechanisms for killing cancer cells (reviewed by Debatin et al., 2002; Reed, 2008) . Thus, Bcl-2 operates at a distal point in a conserved cell death pathway utilized by most anticancer drugs, constituting a form of intrinsic chemoresistance, distinct from the previously identified mechanisms involving drug efflux, drug metabolism, drug inactivation and related mechanisms. This observation presumably explains why expression of a variety of Bcl-2-family proteins has been shown to be of prognostic significance for many types of cancer and leukemia treated by chemotherapy.
Defects in the expression of proapoptotic members of the BCL-2 family also occur in cancer, resulting in loss of the tumor suppressor function of these killer genes. The best documented is BAX, where homozygous deletions or inactivating mutations have been identified, particularly in cancers that arise with microsatellite instability because of defective DNA mismatch repair. In this regard, the human BAX gene contains a homopolymeric stretch of eight guanosine residues in the sense strand that is a target for frame-shift mutations. Defective expression of proapoptotic BCL-2-family genes also occurs in the setting of loss of p53 function. Among the direct targets of the p53 transcription factor are BAX, BID, PUMA and NOXA, thus demonstrating strong connections between genome surveillance by p53 and cell death genes of the BCL-2 family. More recently, cytosolic interactions of p53 protein with pro-and antiapoptotic Bcl-2-family proteins have been observed, directly modulating the bioactivities of the p21-Bax and p26-Bcl-2 proteins, and suggesting that p53 regulates the Bcl-2 family at both transcriptional and post-transcriptional levels. The activities of additional proapoptotic members of the Bcl-2 family are also suppressed through post-translational modifications. For example, proapoptotic protein BAD is phosphorylated by Akt (PKB) and other protein kinases known to be hyperactive in cancers, resulting in its sequestration by 14-3-3 (Oncogene (2008) 27, 6398–6406) .
Overexpression of Bcl-2 prevents apoptosis induced by most and chemotherapeutic drugs, including alkylating agents, and topoisomerase inhibitors. Also, fibroblasts deficient in both Bax and Bak are resistant to apoptosis induced by various agents that induce mitochondrial stress. However, fibroblasts with single deficiency for either Bax or Bak are not significantly defective for apoptosis induced by these agents, suggesting that both Bax and Bak proteins may
be functionally redundant. This apparent redundancy may be cell type-dependent as Bax-deficient HCT116 cells are resistant to apoptosis induced by staurosporine (Theodorakis et al., 2002) . A number of anticancer agents, such as arsenic and lonidamine, can directly permeabilize the outer membrane of mitochondria, at least in vitro, and overexpression of Bcl-2 prevents arsenic-and lonidamine-induced mitochondrial permeabilization and apoptosis. Based on the role of Bcl-2 proteins in regulating mitochondrialmembrane permeabilization, and their frequently modified expression in human cancers, Bcl-2 proteins are legitimate targets (antiapoptotic members) or prototypes (proapoptotic members) for inducing apoptosis either by themselves or in association with other anticancer drugs. However, molecular markers should be used when using Bcl-2-targeted therapies since not all the apoptosis-resistant cells overexpress Bcl-2, as demonstrated in the breast cancer cell lines from the NCI cell screen (Nieves-Neira and Pommier, 1999; and Oncogene (2004) 23, 2934–2949) .
The Bcl-2 protein is a suppressor of programmed cell death that homodimerizes with itself and forms heterodimers with a homologous protein Bax, a promoter of cell death. Bax/Bcl-2 ratio can act as a rheostat which determines cell susceptibility to apoptosis. The subcellular location of some Bcl-2 family members also appears to regulate their function. Bcl-2 and a fraction of Bcl-xL can be found on the mitochondrial outer membrane. In contrast, Bax exists predominantly in the cytosol before apoptosis induction. Early during apoptosis, Bax translocates from the cytosol to mitochondria where it participates in mitochondrial disruption and the release of cytochrome c. Modulating the insertion step can regulate apoptosis. Lower levels of this ratio may lead to resistance of human cancer cells to apoptosis. Thus, Bax/Bcl-2 ratio can affect tumor progression and aggressiveness (Iran Biomed J. 2015 Apr; 19 (2) : 69–75) .
As in the extrinsic pathway, mediators of the intrinsic pathway involved both in tumorigenesis and chemo-resistance are targeted for therapeutic approaches. These anticancer strategies attempt to develop drug-designed inhibitors of anti-apoptotic proteins typically overexpressed in cancer cells, such as Bcl-2, Bcl-xL and IAPs. Efforts to target Bcl-2 proteins involve the development of agents that disrupt Bcl-2 complexes. BH3 mimetics bind to the hydrophobic groove of antiapoptotic proteins mimicking the action of BH3-only proteins in binding to pro-survival proteins, leading to the release of BH3-only proteins from complexes and activation of BAX and BAK. So far, nearly a dozen BH3 mimetics are under investigation as Bcl-2 inhibitors in different phases of human clinical trials such as AT-101 (R- (-) -gossypol) ,
ABT-199 (venetoclax) , ABT-737, ABT-263 (navitoclax, orally available derivative of ABT-737) , GX15-070 (obatoclax) and TW37. The field of inhibitors of Bcl-2 family members is in continuous development, underscoring the importance of these molecules as potent anticancer agents. Moreover, targeting the specific BH4 domain of Bcl-2 is also emerging as a novel strategy for anticancer therapy. Thus, Bcl-2, via its BH4 domain, cooperates with numerous proteins regulating different cellular pathways involved in tumor progression and chemoresistance such as hypoxia and angiogenesis. Recently, a small molecule namely BDA-366 was discovered as a potent and effective BH4 domain antagonist, displaying remarkable anticancer activity in vitro and in vivo, thus providing the proof-of-concept of this approach. Another innovative approach to inhibit Bcl-2 comes from the evidence that human bcl-2 gene contains a GC-rich sequence located in its promoter with the potential to form G-quadruplex structures and functions as a transcriptional repressor element. Therefore, G-quadruplex-specific ligands can regulate the transcription of bcl-2 through stabilization of quadruplex structure (AGING, April 2016, Vol. 8 No. 4) .
Bcl-XL is a major anti-apoptotic protein in the Bcl-2 family whose overexpression is more widely observed in human lung cancer cells than that of Bcl-2, suggesting that Bcl-XL is more biologically relevant and therefore a better therapeutic target for lung cancer. Here, we screened small molecules that selectively target the BH3 domain (aa 90–98) binding pocket of Bcl-XL using the UCSF DOCK 6.1 program suite and the NCI chemical library database. Two new Bcl-XL inhibitors (BXI-61 and BXI-72) that exhibit selective toxicity against lung cancer cells were compared with normal human bronchial epithelial cells. Fluorescence polarization assay reveals that BXI-61 and BXI-72 preferentially bind to Bcl-XL protein but not Bcl2, Bcl-w, Bfl-1/A1 or Mcl-1 in vitro with high binding affinities. Treatment of cells with BXI-72 results in disruption of Bcl-XL/Bak or Bcl-XL/Bax interaction, oligomerization of Bak and cytochrome c release from mitochondria. Importantly, BXI-61 and BXI-72 exhibit more potent efficacy against human lung cancer than ABT-737 but less in platelet reduction in vivo. BXI-72 overcomes acquired radioresistance of lung cancer. Based on our findings, the development of BXI(s) as a new class of anticancer agents is warranted and represents a novel strategy for improving lung cancer outcome (Cancer Res. 2013 Sep 1; 73 (17) : 5485–5496) .
Bax, a central death regulator, is required at the decisional stage of apoptosis. A study recently identified serine 184 (S184) of Bax as a critical functional switch controlling its
proapoptotic activity. Here we used the structural pocket around S184 as a docking site to screen the NCI library of small molecules using the UCSF-DOCK programme suite. Three compounds, small-molecule Bax agonists SMBA1, SMBA2 and SMBA3, induce conformational changes in Bax by blocking S184 phosphorylation, facilitating Bax insertion into mitochondrial membranes and forming Bax oligomers. The latter leads to cytochrome c release and apoptosis in human lung cancer cells, which occurs in a Bax-but not Bak-dependent fashion. SMBA1 potently suppresses lung tumour growth via apoptosis by selectively activating Bax in vivo without significant normal tissue toxicity. Development of Bax agonists as a new class of anticancer drugs offers a strategy for the treatment of lung cancer and other Bax-expressing malignancies (NATURE COMMUNICATIONS DOI: 10.1038/ncomms5935) .
BID (BH3-interacting domain death agonist) protein is situated in extrinsic apoptotic signaling between death receptors and mitochondria, and acts as an inductor of permeabilization of the outer mitochondrial membrane in type II cells. The level of BID is critical for viability of numerous cells because its silencing makes them resistant to apoptosis induced by death receptor ligands, e.g. TNF-related apoptosis-inducing ligand (TRAIL) . Moreover, it has been demonstrated that the level of BID is below the functional dose in cells of several lines because they may be sensitized to TRAIL by overexpression of BID. Due to the above, BID has been considered for therapeutic exploitation. However, to define a way to administer BID, several significant issues should be solved. The main one is control of the level of BID delivered to the cell. Although a full-length BID has been shown to participate in apoptotic signaling, efficient activation of apoptosis needs a specific cleavage of BID by caspase 8 and production of an active truncated form (tBID) . tBID expressed in cells directly induces apoptosis. Therefore, to exploit selectivity for cancer cells exhibited by some anticancer agents, sensitization of cells by full-length BID is a preferred strategy. To sensitize cells, overexpressing systems based on the adenovirus or pcDNA vectors have been commonly used. However, they do not provide stringent control of the level of BID expression in the cell. As a result, the level of BID in transfected cells exceeded several fold that of the endogenous protein and in some cases direct activation of apoptosis was observed instead of sensitization of cells to apoptotic stimuli. According to the above, tBID appears in cells treated with adenovirus vector expressing the full-length BID (BMC Cancer201414: 771 DOI: 10.1186/1471-2407-14-771) .
Rapamycin and its derivatives are promising therapeutic agents with both immunosuppressant and anti-tumor properties. These rapamycin actions are mediated through the specific inhibition of the mTOR protein kinase. It is known that mTOR serves as part of an evolutionarily conserved signaling pathway that controls the cell cycle in response to changing nutrient levels. The mTOR signaling network contains a number of tumor suppressor genes including PTEN, LKB1, TSC1, and TSC2, and a number of proto-oncogenes including PI3K, Akt, and eIF4E, and mTOR signaling is constitutively activated in many tumor types. These observations point to mTOR as an ideal target for anti-cancer agents and suggest that rapamycin is such an agent. In fact, early preclinical and clinical studies indicate that rapamycin derivatives have efficacy as anti-tumor agents both alone and when combined with other modes of therapy. Rapamycin appears to inhibit tumor growth by halting tumor cell proliferation, inducing tumor cell apoptosis, and suppressing tumor angiogenesis. Rapamycin immunosuppressant actions result from the inhibition of T and B cell proliferation through the same mechanisms by which rapamycin blocks cancer cell proliferation. Therefore, one might think that rapamycin-induced immunosuppression would be detrimental to the use of rapamycin as an anti-cancer agent. To the contrary, rapamycin decreases the frequency of tumor formation that occurs in organ transplant experiments when combined with the widely used immunosuppressant cyclosporine compared with the tumor incidence observed when cyclosporine is used alone. The available evidence indicates that with respect to tumor growth, rapamycin anti-cancer activities are dominant over rapamycin immunosuppressant effects. In recent years, clinically rapamycin and its analogs (like CCI-779 or temsirolimus, RAD001 or everolimus, Sirolimus, FK-50 and AP23576) have been used in the treatment of various cancers, including kidney cancer, mantle cell lymphoma and metastatic breast cancer.
Dipyridamole (DPM) , like aspirin, inhibits platelet adhesion, and thus tends to prevent the vascular thrombosis of heart attacks and strokes. In the December 12, 1987 issue of the Lancet (pp. 1, 371-4) was a report on the European Stroke Prevention Study. The introduction to this report reviewed the indicated lack of benefits in treating patients with aspirin who had survived a small stroke, a TIA, a temporary ischemic attack. In this trial, dipyridamole 300 mg a day was added to treatment with aspirin and the results were outstanding. Over a two-year period, stroke deaths were decreased by 50%, deaths from myocardial infarction decreased by 38%and deaths from cancer by 25%. The above-indicated anti-cancer effect of dipyridamole
may be due only to its prevention of metastases; however, Eva Bestida et al. of the University of Barcelona had a report in Cancer Research, the September 1985 issue (pp. 4, 048-4, 062) on the inhibition of certain human cancer cell growths by dipyridamole. It caused an inhibition of greater than 80%of adinosine, thymidine and uridine. These are substances needed by cancer cells to prosper. This may indicate an anti-cancer effect of dipyridamole other than in the prevention of metastases.
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has emerged as an attractive cytokine that selectively targets cancer cells; however, its efficacy has been challenged by a number of resistance mechanisms. Therefore, the current study investigated the potential of dipyridamole to enhance TRAIL efficacy and the probable underlying mechanisms. Dipyridamole dramatically sensitized p53-mutant human cancer cell lines: SW480, MG63 and DU145, to the antitumor activity of TRAIL, as evidenced by enabling TRAIL to efficiently cleave initiator and executioner caspases. Although dipyridamole upregulated both DR4 and DR5 and increased their cell surface expression, RNA interference revealed a preferential dependence on DR5. Moreover, dipyridamole inhibited survivin expression and its important consequences were confirmed by small interfering RNA. Mechanistically, dipyridamole induced transcriptional shutdown of survivin expression accompanying G1 arrest that was characterized by downregulation of D-type cyclins and cdk6. In addition, a transcriptional mechanism powered by CCAAT/enhancer-binding protein (C/EBP) homologous protein (CHOP) induction was responsible for DR5 upregulation by dipyridamole. Importantly, dipyridamole-induced enhancement of TRAIL efficacy and alterations of protein expression were independent of either protein kinase A or protein kinase G. In conclusion, findings of the present study describe novel mechanisms of dipyridamole action and highlight its promising use as a potential enhancer of TRAIL efficacy (Oncogene (2008) 27, 3435–3445) .
Preliminary observations indicate that Dipyridamole (DPM) can increase the sensitivity of human ovarian carcinoma cells to cisplatin. In another investigation, Jekunen et al., showed that DPM synergistically enhanced the cytotoxicity of cisplatin in cisplatin-sensitive 2008 human ovarian carcinoma cells by a factor of 4.7, and in the cisplatin-resistant 2008/C13*5.25 subline by a factor of 5.8. DPM was found to increase the cellular uptake of cisplatin in a concentration dependent manner, without increasing trypan blue or propidium iodide uptake or changing cell size. They concluded that the DPM-induced increase in cisplatin
accumulation was not associated with a nonspecific increase in membrane permeability. In a nude mouse model with human bladder cancer, Keane et al., found that tumor size decreased by 20%when cisplatin was combined with DPM. Using human testicular carcinoma in the same model, they achieved complete tumor regression. Barberi-Heyob et al. found that DPM synergistically increased the growth-inhibitory activity of cisplatin in MCF-7 human breast cancer cells. Janice R. Perussi et al. reported here that the enhancement of cisplatin cytotoxicity by DPM in cisplatin-sensitive MDA/Shuman breast cancer cells suggest that there is a strong correlation between Pt accumulation and enhanced cisplatin cytotoxicity, but in cisplatin-resistant MDA/R cells the synergistic interaction between cisplatin and DPM does not involve an increase in the cellular accumulation of cisplatin (Quím. Nova vol. 26 no. 3 Paulo May/June 2003) .
Gemcitabine (2’, 2’-difluorodeoxycytidine) is a pyrimidine nucleoside analogue. It exerts its cytotoxic effect intracellularly and has activity against a number of different solid tumors, including pancreatic, breast, lung, and bladder cancers. As gemcitabine is strongly hydrophilic, passive diffusion through the hydrophobic cellular plasma membrane lipid bilayer is slow. In order to efficiently enter cells, gemcitabine requires physiologic nucleoside transporter proteins to cross the plasma membrane. These transporter proteins fall into two categories, equilibrative transporters and concentrative transporters. The bi-directional human equilibrative nucleoside transporters (hENT) are found in most cell types, and both hENT1 and hENT2 are capable of mediating gemcitabine uptake in the direction of the concentration gradient. The hENT proteins are transmembrane glycoproteins that localize to the plasma membrane. They are functionally distinguished by their ability to be inhibited by nitrobenzylmercaptopurine ribonucleoside (NBMPR) , a specific hENT1 inhibitor at low nanomolar concentrations, and dipyridamole, a hENT1/2 inhibitor, with reduced sensitivity to gemcitabine by 39-and 1, 800-fold (The Oncologist 2008; 13: 261–276) .
Fluorouracil (5-FU) , sold under the brand name Adrucil among others, is a medication used to treat cancer. By injection into a vein it is used for colon cancer, esophageal cancer, stomach cancer, pancreatic cancer, breast cancer, and cervical cancer. As a cream it is used for actinic keratosis and basal cell carcinoma and as eye drops for treatment of ocular surface squamous neoplasia.
Defects in apoptosis have been implicated in chemoresistance of cancer cells. Studies have found that high levels of anti-apoptotic Bcl-2 combined with a low level of Bax were correlated to high 5-FU resistance in various cancers, including human breast adenocarcinoma, pancreas carcinoma, human head and neck carcinoma, colon tumor cells (Int J Cancer. 2002 Apr 1; 98 (4) : 498-504; and Br J Cancer. 2000 Nov; 83 (10) : 1380–1386) .
Dipyridamole has been shown to potentiate the cytotoxicity of 5-FU and other fluoropyrimidines in vitro. This effect probably relates to increases in intracellular concentrations of FdUMP and to the inhibition of the uptake of extracellular nucleosides by the "salvage" pathway. In vitro studies have shown that dipyridamole can augment the cytotoxicity of a number of cytotoxic agents, including etoposide, doxorubicin, vinblastine, and mitoxantrone, perhaps by altering cellular uptake and retention of the cytotoxic agents (Investigational New Drugs 12: 283-287, 1994) .
Multidrug resistance (MDR) in cancer cells is the simultaneous development of resistance to a variety of anticancer drugs that appear to be structurally and mechanistically unrelated. One type of MDR is characterized by the decreased accumulation of hydrophobic natural product drugs. In some multidrug-resistant cells, drug efflux is mediated by an adenosine triphosphate (ATP) -dependent membrane transporter termed P-glycoprotein (Pgp) , the product of the MDR1 gene (Juliano and Ling, 1976) . Pgp functions as an active outward transport mechanism for a variety of molecules, including certain chemotherapeutic drugs. As data accumulated regarding the role of Pgp in drug resistance, it became clear that other transporters could confer resistance to cytotoxic agents. The MDR protein 1 (MRP1) gene was cloned from a multidrug-resistant lung cancer cell line and, like Pgp, was found to be a member of the ATP-binding cassette (ABC) superfamily of transporter genes (Cole et al., 1992) . Transfection studies indicated that, similar to MDR1, MRP1 overexpression is sufficient to confer resistance to a broad profile of lipophilic, natural product antineoplastics. MRP1 was the first identified member of a family of genes encoding multispecific organic anion transporter (MOAT) proteins (Borst et al., 1999) . Two other homologues of MRP1, the cMOAT/MRP2 and MRP3 genes, encode proteins that mediate MDR when transfected into a drug-sensitive cell (Borst et al., 1999) . All of these membrane-embedded proteins act as drug efflux pumps, preventing cytotoxic agents from reaching lethal levels within cells.
P-glycoprotein (P-gp) is a key player in the multidrug-resistant phenotype in cancer. The protein confers resistance by mediating the ATP-dependent efflux of an astonishing array of anticancer drugs. Its broad specificity has been the subject of numerous attempts to inhibit the protein and restore the efficacy of anticancer drugs. The general strategy has been to develop compounds that either compete with anticancer drugs for transport or act as direct inhibitors of P-gp. Despite considerable in vitro success, there are no compounds currently available to “block” P-gp-mediated resistance in the clinic. The failure may be attributed to toxicity, adverse drug interaction, and numerous pharmacokinetic issues. In addition, multidrug resistance-associated protein 1 (MRP1) transports a wide range of therapeutic agents as well as diverse physiological substrates and may play a role in the development of drug resistance in several cancers, including those of the lung, breast and prostate, as well as childhood neuroblastoma. Several studies above have shown that in vitro, dipyridamole can significantly increase the cytotoxic and antitumor activities of a variety of chemotherapeutic agents. The underlying mechanism here is both prevention of nucleoside and nucleobase salvage, and an increase in the intracellular accumulation of the toxic metabolites via inhibited P-glycoprotein and MRP1 (Clin Pharmacol Ther 2003; 73: 51–60. Drug Metab Dispos. 2014 Apr; 42 (4) : 623–631. Oncogene (2003) 22, 7340–7358) .
The study has investigated the potential role of dipyridamole as a single agent in the prevention of tumorigenesis and metastasis in multiple models of triple negative (estrogen and progesterone receptor-negative, Her-2 normal) breast cancers, a subtype that has few effective therapies. These findings provide evidence that intraperitoneal administration of dipyridamole impairs primary tumor growth and metastasis in breast-cancer xenograft animal models. Moreover, our data identify new mechanisms of action of dipyridamole, which is shown to inhibit the ERK1/2-MAPK, NF-kB and Wnt signaling pathways, and to prevent the accumulation of inflammatory cells in the tumor microenvironment. In addition, dipyridamole was the most potent BCRP inhibitor among the compounds tested with IC50 values of 6.4 +/-0.9 microM. Therefore, dipyridamole has the potential to treat cancer multidrug resistance (Cancer Prev Res (Phila) . 2013 May ; 6 (5) : . doi: 10.1158/1940-6207. CAPR-12-0345) .
However, clinically administered DPM did not improve the anticancer activity of 5-FU or cisplatin in patients with advanced colorectal cancer, metastatic breast cancer, advanced
non-small cell lung cancer or advanced measurable pancreatic cancer. The observed increase in 5-FU or cisplatin dose-intensity for DPM was not clinically relevant.
The way nanoparticle drug carriers enter cells is different from that of conventional drugs. Conventional drugs enter cells by diffusion, which is dose-dependent. That is, the higher the drug concentration in the blood, the higher the drug concentration in the cells, and the drugs can only enter cytoplasm. Nanoparticle drug carriers are absorbed by cells through endocytosis and are lysosomotropic after entering cells. At the initial stage after injection, the concentration of the nanoparticle drug carriers increases in a time-dependent manner.
Endocytosis is a process to incorporate extracellular materials into cells. This process can be categorized into three types, i.e., phagocytosis, pinocytosis, and receptor-mediated endocytosis. Phagocytosis only occurs in specialized cells. These cells proliferate and aggregate upon stimulation by extracellular materials and engulf them into lysosomes in the cells for degradation. This process occurs in macrophages and neutrophils of the immune system. Pinocytosis is a process that internalizes extracellular fluid and molecules within it through the invagination of the cell membrane to form a pocket, which then pinches off into the cell to form a vesicle. The vesicle then travels into the cytosol and fuses with other vesicles such as endosomes and lysosomes.
Depending on the structure of the carriers, pinocytosis can be categorized into two types, fluid phase pinocytosis and adsorptive pinocytosis. If the carrier does not have a functional group that interacts with the cells, the cells will engulf the drug carrier by fluid phase pinocytosis. This process is slow and dependent on the carrier concentration around the cell membrane. Adsorptive pinocytosis occurs when the carrier has a hydrophobic group or is positively charged. Such carrier will be physically adsorbed by the cell membrane and increase the engulfing ability of the cells. The above two types of endocytosis are non-specific processes and are not suitable for delivery of drugs to their targets. Targeting can only be achieved in certain cancer tissues through enhanced permeability and retention (EPR) .
Receptor-mediated endocytosis is a process by which cells absorb molecules (endocytosis) by the inward budding of plasma membrane vesicles containing proteins with receptor sites specific to the molecules being absorbed. After the drug carrier binds to the receptor on the cell, an intrinsic signal will trigger the cell membrane to form a coated pit. The surface area of a coated pit amounts to 1 to 2%of the cell membrane. The coated pit will detach
from the cell membrane and enter into the cell to form coated vesicles in the cell, and subsequently form endosomes and move inside the cell in saltatory motion. An endosome is a complicated structure comprising microtubules and vesicles. The vesicles can fuse with Golgi. Due to the proton pump (ATPase) , endosomes usually become acidic. The endosomes will then fuse with lysosomes to form secondary lysosomes.
The cell membrane is a barrier to be overcome for efficient delivery of therapeutics into a target site in mitochondria, cytoplasm or nucleus. Hydrophobic phospholipids are major components of the cell membrane that obstruct the transportation of therapeutics. Thus, various delivery systems, such as liposomes, nanoparticles and viral vectors, have been developed to transfer small molecules, peptides, proteins, and oligonucleotides across the membrane. Such manner of drug delivery is herein referred to as cell-penetrating drug delivery systems.
A number of drug carrier systems (liposomes, cell penetrating peptides, cationic polymer conjugates, and polymeric nanoparticles) have been explored for intracellular delivery of therapeutics. They need to be adapted to cross a series of membrane barriers in order to reach the site of drug action in the cells. During this process, a significant portion of the drug molecules will be lost at each successive barrier. These barriers include cellular association and internalization of the drug-carriers by endocytosis; intracellular trafficking and release of drug or drug-carrier into the cytoplasm; cytoplasmic translocation of drug or drug-carrier to nucleus or any other cellular organelle; and the nuclear/organellar uptake. Cells contain several intracellular organelles with specific functions. Intracellular targeting of therapeutics to these specific organelles is expected not only to significantly enhance the therapeutic efficacy but also reduce non-specific effect and hence toxicity. Therefore, there is significant interest in achieving intracellular target-specific delivery of therapeutics using different carrier systems.
The carriers that facilitate the endocytosis of drugs include nano-sized polymeric carriers and liposomes. Depending on the properties of the drugs and preparation processes, nano-sized drug carriers can be categorized into nanoparticles, nanoliposomes, nano suspended particles, solid lipid nanoparticles, magnetic nano-carriers, and the like.
In addition to the above-mentioned carriers, cell-penetrating peptides (CPP) , biodegradable nanoparticles, and viral vectors may also be used as delivery systems for enhancing the penetration of drugs into cells.
As a cell membrane constitutes a major barrier for intracellular delivery of large-sized hydrophilic proteins, peptides and oligonucleotides, cell penetrating peptides (CPPs) have been explored to overcome this barrier. These CPPs can ferry molecules or colloidal drug carrier systems that are tagged to them across the cell membrane, into the cytoplasm and to the nucleus. The characteristics of CPPs are attributed to the presence of a stretch of 9-16 cationic amino acid residues; the most commonly studied CPPs include HIV-1 transactivating transcriptional activator (TAT) peptide, HSV VP-22 (Herpes Simplex virus type-1 transcription factor) peptide and penetratin. Several theories have been proposed to determine the exact mechanism by which these CPPs enter the cells. For example, TAT penetration through cell membrane has been shown to be independent of receptors and transporters, and has been suggested to enter the cell by forming an inverted micelle by destabilizing the phospholipid bilayer. The main benefit of TAT coupling is that, along with efficient delivery of molecules, biological activity of the coupled molecule is preserved, and the size of the molecule being transported is also not a rate-limiting factor.
TAT has been suggested not only to enhance intracellular delivery, but also nuclear delivery, and hence has been investigated for nucleic acid delivery. TAT peptide conjugated to antisense oligonucleotide has been shown to deliver oligonucleotides to the nucleus. After being internalized, TAT peptide has also been found to co-localize inside the Golgi body along with BODIPY-ceramide, which is a marker for Golgi body. Therefore, it is quite possible that there is direct trafficking from the early endosome to the Golgi body without entering the late endosome. A secretory pathway could be present where the peptide enters the cytosol from the endoplasmic reticulum. Gene therapy has demonstrated significant potential in the treatment of genetic, acquired and neurodegenerative disorders. Amongst non-viral gene delivery methods, various drug delivery systems and polymers are being investigated such as liposomes, cationic lipid-DNA, polymer complexes. To overcome relatively inefficient cellular uptake of non-viral gene expression vectors, TAT peptide conjugation to vectors has been explored.
Kleeman et al. have demonstrated gene expression in alveolar basal epithelial cells with polyethylenimine (PEI) covalently coupled to TAT through a polyethylene glycol (PEG) spacer, which demonstrated higher transfection efficiencies in vivo in mice lung following intratracheal administration than unconjugated PEG complex. In a similar study by Rudolph et
al., solid lipid particles conjugated to dimeric HIV-1 TAT have demonstrated enhanced gene delivery to the lungs.
CPPs typically have an amino acid composition that either contains a high relative abundance of positively charged amino acids such as lysine or arginine or has sequences that contain an alternating pattern of polar/charged amino acids and non-polar, hydrophobic amino acids. These two types of structures are referred to as polycationic or amphipathic, respectively. A third class of CPPs are the hydrophobic peptides, containing only apolar residues, with low net charge or having hydrophobic amino acid groups that are crucial for cellular uptake. Among the cell-penetrating peptides, the arginine-rich cell-penetrating peptides have been the most widely studied. Examples include the TAT peptide from the HIV transactivator protein TAT, Penetratin, a 16 amino acid domain from the Antennapedia protein of Drosophila, a flock house virus (FHV) coat peptide (sequence 35–49) , and oligoarginines.
Biodegradable nanoparticle-mediated intracellular delivery is a dynamic process; involving endocytosis, exocytosis, and sorting into different intracellular compartments. It appears that the NP surface and its interaction with cell surface controls the uptake and intracellular trafficking of biodegradable nanoparticles, and hence that of the encapsulated therapeutic agents.
Viral vectors are tools commonly used by molecular biologists to deliver genetic materials into cells. This process can be performed inside a living organism (in vivo) or in cell culture (in vitro) . Hence, viral vectors are applicable options for use in cell-penetrating drug delivery systems.
Cell-penetrating peptides and biodegradable nanoparticles are used not only to modify drugs but also to be conjugated to carries to enhance the transmembrane effects.
Dipyridamole is an equilibrative nucleoside transporter (ENT) inhibitor. Nucleoside transporters (NTs) play an essential role in the transport of nucleosides across cellular membranes. Dipyridamole blocks the equilibrative nucleoside transporter (ENT) , which facilitates the transmembranous diffusion of adenosine. Dipyridamole will increase the extracellular endogenous adenosine concentration, mainly in situations of increased extracellular formation of adenosine, such as occurs during hypoxia or inflammation. However, the extracellular endogenous adenosine concentration induced by dipyridamole causes vasodilatation, which contributes to the metabolic control of organ perfusion. Dipyridamole stress myocardial
imaging is a widely used and successful technique for diagnosing and evaluating coronary artery disease. Coronary vasodilation with IV dipyridamole is associated with significant reductions in blood flow to collateral-dependent myocardium consistent with coronary steal in patients with CAD. In addition, there have been further studies that discovered vasoconstrictor and vasodilator effects of dipyridamole in many organs, including kidney, lung, pancreas, brain and so on.
Dipyridamole not only causes vasoconstriction in some organs but can also lead to low blood pressure and subsequent side effects such as vertigo and palpitations due to dilation of blood vessels of the heart. The effect of reducing blood pressure makes dipyridamole unsuitable for the treatment of patients who are physiologically unstable, such as those having, but not limited to, sepsis, ischemic stroke, hemorrhagic stroke, acute lung injury, acute liver injury, myocardium infarct, and cardiorenal syndrome. Furthermore, the blood-flow restricting effect of dipyridamole limits its application in the treatment of diseases involving organs rich with blood vessels.
Since the pharmacological action of dipyridamole is mainly on cell membranes, a delivery system designed for membrane penetration avoiding binding with equilibrative nucleoside transporter on the membrane while enhancing the intracellular signal transduction and PPARγ regulation can prevent the effect of tissue hypoperfusion due to increased cardiovascular dilation and local blood flow restriction. The limitation in clinical applications of dipyridamole in acute and severe patients due to the decrease of blood pressure can thus be lifted.
Dipyridamole is also a non-selective phosphodiesterase inhibitor. Increase of intracellular drug delivery will enhance the inhibition of dipyridamole on intracellular phosphodiesterase (PDE) . Members of the PDE family have unique cell-and tissue-specific distribution. Dipyridamole may be used as anti-inflammatory, anti-oxidant, anti-fibrosis, and smooth muscle relaxing agents for treating diseases associated with PDE regulation depending on the distribution profile of PDE on cell membranes or in cytoplasm in different tissues.
The unique cell-and tissue-specific distribution of PDEs are shown in the table below (see US 2012/0065165) :
PDE
Increase on the capability of dipyridamole to penetrate the membrane can facilitate the inhibition of PDE3, PDE5 and PDE8 in specific tissues and confer dipyridamole therapeutic efficacy in diseases associated with PDE3, PDE5 and PDE8. In such case, dipyridamole may be used for treating lower urinary tract dysfunction and erectile dysfunction, like other PDE5 inhibitors. Furthermore, since dipyridamole is a non-selective PDE inhibitor, it may be used for the treatment of PDE associated diseases when delivered by a transmembrane drug delivery system.
There are numerous molecules currently in use or being tested in clinical trials that act on mitochondria. Several clinically approved anticancer drugs such as paclitaxel and VP-16 (etoposide) and vinorelbine as well as an increasing number of experimental anticancer drugs such as ceramide, MKT077, CD437, lonidamine, and betulinic acid have been found to act directly on mitochondria to trigger apoptosis. CD437 is able to induce apoptosis in a variety of human carcinoma cells in vitro and in vivo. In intact cells, CD437-dependent caspase activation
is preceded by the release of cytochrome C from mitochondria. Moreover, when added to isolated mitochondria, CD437 causes membrane permeabilization. This effect is prevented by inhibitors of the mitochondrial permeability transition pore complex (mPTPC) , such as cyclosporine A. Therefore, CD437 represents a low molecular weight compound which exerts its cytotoxic effect via the mPTPC, i.e., by acting directly at the surface or inside of mitochondria. Similarly, arsenic trioxide, which is used in the treatment of acute promyelocytic leukemia, has multiple actions on mitochondria. It is known to cause the induction of mPTPC formation via its action on the voltage-dependent anion channel VDAC. Arsenic trioxide is also known to act on the respiratory chain and inhibit respiratory chain activity. Apoptotic factors that play a major role in the modulation of apoptosis include Bcl-2 and Bcl-Xl. Compounds that act by binding to these proteins have been identified and studied for their efficacy; a few examples include a chromene derivative and gossypol which was recently shown to act on proteins of the Bcl-2 family. In fact there are so many varied and structurally different compounds that it has been suggested that they be collectively called mitocans to reflect their mitochondrially mediated anticancer effects.
The selective accumulation approach to targeting tumor mitochondria requires two levels of specific accumulation; drug accumulation in the tumor and then drug accumulation in the mitochondria of cancer cells. Generally speaking, drug disposition may be modulated by subtle modification of the chemical structure to change its physico-chemical properties that are known to determine its accumulation in various compartments. Of course such modification must be done without adversely affecting action on the molecular target. The second approach involves conjugating ligands that are larger than simple organic functional groups to change the biodistribution of the active molecule. Again this approach works as long as the conjugation does not adversely affect the desired pharmacological activity of the molecule. Such approaches have been used very effectively to alter drug distribution in the body and achieve higher accumulation in target tissues using ligands that are known to have an affinity for the target tissue. There are ligands that have been shown to mediate tumor-specific accumulation of drugs, and there are ligands that are known mitochondriotropics. However, it is still unclear whether there exists a ligand that possesses both properties to a degree that will allow high levels of the desired accumulation. It may therefore be safe to say that, for now, a dual strategy is the most feasible approach. Such a dual strategy would require the use of one targeted delivery approach
to achieve high tumor accumulation followed by a second approach to ensure that the drug then accumulates in the mitochondria where it will exert its action. While there is much investigation into tissue-specific delivery aimed at increasing tumor levels of anticancer drugs, research aimed at sub-cellular delivery is only just gaining greater attention. Nonetheless there are some interesting approaches to mitochondrial delivery that suggest the promise of improved therapy for cancer.
Pharmaceutical nanocarriers like liposomes, micelles, and solid nanoparticles offer what might be viewed as a non-chemical approach to modify the disposition of drug molecules. All chemistry can be performed on the components of the nanocarrier system that can then be loaded with the drug to afford targeted delivery. Most pharmaceutical nanocarriers can be modified for targeting to specific tissues and even specific cell types. Long-circulating liposomes and nanoparticles are able to passively target areas of leaky vasculature by virtue of the enhanced permeability and retention (EPR) effect and can additionally be modified with antibodies or other targeting ligands to afford cell-specific recognition. Nanocarriers that can not only affect the tumor-specific accumulation of a drug but also mediate mitochondria-specific accumulation within a tumor cell might be the ultimate tool in mitochondria-targeted anticancer approaches if they can be developed for clinical therapy. The first steps in this direction have already been taken in recent years. Current nanocarrier technology is reaching the point where the need for sub-cellular delivery may indeed be met using nanocarriers specifically designed for such purposes.
In an approach towards the delivery of the mPTPC inducing drug mastoparan into cells, liposomes were modified with both transferrin and a fusogenic peptide Chol-GALA. The transferrin modification enhances liposomal uptake into cells via endocytosis after which the peptide facilitates release from the endosomes into the cytosol. Thus, by just increasing the intracellular content of the drug, the delivery approach achieved a higher concentration of the drug that was potentially available to interact with the sub-cellular target. Micelles have also been proposed for the delivery of hydrophobic drugs to various sub-cellular organelles including mitochondria. The fluorescently labeled micelles used in the study were found to be distributed through several cytoplasmic organelles, including a majority of them associated with the mitochondria. The uptake of these micelles was not restricted to a single cell type. Also, the extent of cell internalized cargo incorporated in micelles was greater than the free cargo by itself.
There are now several examples of nanocarriers designed specifically to accumulate in mitochondria. Arguably the earliest of these are what are known as DQAsomes. Prepared from the mitochondriotropic molecule dequalinium chloride, these vesicular nanocarriers were developed for mitochondria-specific DNA delivery but were also shown to be capable of changing the sub-cellular distribution of paclitaxel to increase the accumulation of the drug in mitochondria. The mitochondria-specific delivery led to improved apoptotic activity at paclitaxel concentrations, at which the free drug does not have a significant cytotoxic effect. Paclitaxel loaded DQAsomes have also been tested for their ability to inhibit the growth of human colon cancer tumors in nude mice and the data strongly suggest that encapsulation of paclitaxel in DQAsomes leads to improved efficacy. The antitumor efficiency of DQAsomal encapsulated paclitaxel was also further enhanced by modifying the DQAsomal surface with folic acid (FA) . The folate receptor is a folate high-affinity membrane binding protein, which is overexpressed in a large variety of human tumors. FA conjugates are internalized in a tumor cell-specific manner by receptor-mediated endocytosis resulting in an increased toxicity of the corresponding drug.
Another approach to the design of mitochondria-specific nanocarriers is to modify existing nanocarriers with mitochondriotropic ligands. In this regard TPP again served as the mitochondriotropic ligand in liposomal and polymer based nanocarriers. Liposomes have been well characterized as delivery systems and are a popular choice due to their biocompatibility, ease of surface modification, and capacity to encapsulate hydrophilic or hydrophobic drugs. The first indication that liposomes could be rendered mitochondriotropic by surface modification with a mitochondriotropic residue came from a report on so-called proteoliposomes prepared by incorporating a crude mitochondrial membrane fraction into liposomes colocalized with endogenous mitochondria in pre-implantation embryos. Further investigation of the concept of using ligands to alter the sub-cellular distribution of liposomes was based on the synthesis of stearyltriphenylphosphonium (STPP) . The stearyl residue of STPP serves as a lipid anchor to modify the surface of liposomes with the TPP residue and results in a liposomal preparation with a marked predisposition for mitochondria. STPP-liposomes were shown to effectively direct the accumulation of rhodamine labeled phosphatidylethanolamine (Rh-PE) to mitochondria in live cells. Based on flow cytometry, STPP-liposomes exhibited the same level of cell association as liposomes with the same cationic charge. However, the subsequent sub-cellular localization
analyzed by confocal microscopy was markedly different, indicating that the mitochondriotropic ligand, and not the surface charge, is what determines mitochondria-specific association of the nanocarrier. It was also found that the TPP ligand did not change the in vivo distribution and tumor accumulation of long-circulating PEGylated liposomes. STPP-liposomes did, however, improve the both the in vitro and the in vivo efficacy of ceramide. Taken together these data suggest that the tendency of long-circulating liposomes to passively accumulate (via the EPR effect) in solid tumors can be combined with organelle-specific tropism conferred by modification with an appropriate ligand to potentiate the effect of an encapsulated antitumor agent.
An alternative approach to developing mitochondria-specific liposomes has focused on the concept that liposomes with a tendency to selectively fuse with mitochondrial membranes are more likely to associate with mitochondria upon cell entry. Referred to as MITO-Porter, these liposomes are surface modified with octaarginine residues to facilitate their entry into cells as intact vesicles (via macropinocytosis) . The lipid composition was selected on the basis of high levels of fusion with the mitochondrial membrane and the release of its cargo to the intra-mitochondrial compartment in living cells. Based on confocal microscopy data, MITO-porter liposomes have been used to deliver green fluorescent protein as well as propidium iodide to mitochondria, suggesting that they can be used to deliver macromolecules as well as small molecules to mitochondria.
The development of mitochondria-specific nanocarriers has not been limited to lipid based carriers but also includes the use of mitochondriotropic residues to create polymeric systems capable of mitochondria-specific intracellular delivery of bioactive molecules. TPP modification has been employed to create a mitochondriotropic N- (2-hydroxylpropyl) methacrylamide (HPMA) copolymer-based nanoparticle. Interestingly, earlier study indicated that while the polymers characterized did exhibit association with isolated mitochondria, experiments with ovarian carcinoma cells revealed predominantly lysosomal association of the polymer. However, in a more recent study, microinjection and incubation experiments performed using fluorescently labeled constructs suggested mitochondrial targeting ability based on microscopic analysis. Subsequently, HPMA copolymer-drug conjugates were synthesized using a photosensitizer mesochlorin e 6 (Mce 6) . Mitochondrial targeting of HPMA copolymer-bound Mce 6 enhanced cytotoxicity as compared to non-targeted HPMA copolymer-
Mce 6 conjugates. The authors indicate that “minor modifications may be required to adapt the current design and allow for tumor site-specific mitochondrial targeting of other therapeutic agents. ” Therefore these systems could theoretically be applied to the mitochondria-specific delivery of a range of pro-apoptotic substances for cancer therapy.
Inorganic nanoparticles have also been shown to be capable of mitochondria-specific delivery. In a very recent study, hypocrellin A, a photodynamic drug, was encapsulated in a water-soluble amorphous silica nanocage (HANC) . These drug-loaded nanocages are reportedly able to specifically accumulate in the mitochondria of cancer cells and improve the photosensitizing effect of hypocrelin A. It is, however, unclear what mediates the mitochondria-specific accumulation of the nanocage system. Nevertheless, taken together, the various studies described so far strongly support the hypothesis that nanocarriers can indeed control the sub-cellular accumulation of bioactive molecules and as such represent a useful tool in the development of mitochondria-targeted anticancer strategies (G. G. M. D'S ouza et al. /Biochimica et Biophysica Acta 1807 (2011) 689–696) .
Extracellular adenosine disrupted mitochondrial membrane potentials in HuH-7 cells, a Fas-deficient human hepatoma cell line, and the effect was inhibited by the adenosine transporter inhibitor dipyridamole or by overexpressing Bcl-XL. Adenosine downregulated the expression of mRNAs and proteins for Bcl-XL and inhibitor of apoptosis protein 2 (IAP2) to directly inhibit caspase-3, -7, and -9, but it otherwise upregulated the expression of mRNA and protein for DIABLO, an inhibitor of IAPs. Those adenosine effects were attenuated by dipyridamole (Cell Biol Toxicol (2010) 26: 319–330) .
Extracellular adenosine induces apoptosis in a variety of cancer cells via intrinsic and extrinsic pathways. In the former pathway, adenosine uptake into cells triggers apoptosis, and in the latter pathway, adenosine receptors mediate apoptosis. Dipyridamole inhibiting the cellular uptake of adenosine reversed significantly the adenosine-induced growth suppression.
Summary of the Invention
It is found in the present invention that certain substituted pyrimido [5, 4-d] pyrimidine compounds such as dipyridamole are capable of enhancing the expression and activity of BAX/BCL-2. Therefore, the present invention provides a novel type of BAX/BCL-2 modulator having the pyrimido [5, 4-d] pyrimidine main structure and a method of preventing or
treating BAX/BCL-2-related disorders or conditions, such as cancers, myeloproliferative disorder, prostate epithelial dysplasia, lymphangioleiomyomatosis, Kimura disease, and keloid, using such BAX/BCL-2 modulators. The invention also relates to a method of increasing the expression and activity of BAX/BCL-2.
Although research demonstrates that dipyridamole exhibits anticancer activity, the efficacy is insignificant at low dose. Dipyridamole is a known ENT-1 inhibitor which blocks the uptake of adenosine into the cells. Since adenosine may promote apoptosis of cancer cells, such blockade decreases the anticancer efficacy of dipyridamole. In the present invention, dipyridamole may be encapsulated in a carrier to facilitate penetration of the cell membrane and accumulation in the cells, and thereby activate the endogenous apoptosis mechanism of the cells. The enhanced pro-apoptotic efficacy of dipyridamole in the cells may be achieved through increasing the formation of BAX/BCL-2 heterodimer, increasing the formation of BAX homodimer and decreasing the formation of BCL-2 homodimer. It is also found in the present invention that dipyridamole can reduce BCL-XL expression, reduce the inhibition of ENT-1 and modulate the apoptotic protein expression of the cancer cell, thereby increasing its anticancer ability.
In a preferred embodiment, dipyridamole is encapsulated in a cell-penetrating carrier to increase the concentration of dipyridamole inside the cell and reduce the inhibition on ENT-1. In an embodiment for treating cancer, increase of BCL-XL and decrease of cytosolic adenosine, and thus decrease of anticancer efficacy, can be avoided.
In an embodiment, the invention relates to a method of preventing or treating a BAX/BCL-2-related disorder or condition, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of formula (I) :
wherein each of R1, R2, R3 and R4 is independently selected from the group consisting of heterocyclyl and di (hydroxyalkyl) amino,
or a pharmaceutically acceptable salt thereof.
The present invention also relates to use of a compound of formula I or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for preventing or treating BAX/BCL-2-related disorders or conditions. In a preferred embodiment, the medicament comprises a compound of formula I or a pharmaceutically acceptable salt thereof encapsulated in a pharmaceutically acceptable carrier.
The present invention further relates to a pharmaceutical composition for preventing or treating BAX/BCL-2-related diseases, comprising a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt thereof encapsulated in a pharmaceutically acceptable carrier. In a preferred embodiment, the compound is dipyridamole and the carrier is a liposome.
The present invention is described in detail in the following sections. Other characterizations, purposes and advantages of the present invention can be easily found in the detailed descriptions and claims of the invention.
Figures 1a and 1b are schemes showing the different actions of dipyridamole (a) outside the cell and (b) inside the cell on cancer cell apoptosis.
Figure 2 demonstrates the correlation between cell viability and Bax/Bcl-2 ratio (Y-axis: cell viability; X-axis: Bax/Bcl-2 ratio) .
Figure 3 shows the structures of representative Rapalogs.
Figures 4a and 4b show the cell viability of the triple-negative breast cancer cell line MDA-MB-231 treated by dipyridamole with/without 5-FU.
Figures 5a and 5b show the Bax, Bcl-2 and Bcl-xL expression of cancer cells treated by dipyridamole with/without 5-FU.
Figures 6a and 6b show the Bax/Bcl-2 ratio in cells treated by dipyridamole with/without 5-FU.
Unless otherwise defined herein, scientific and technical terms used in connection with the present invention shall have the meanings that are commonly understood by those of ordinary skill in the art. The meaning and scope of the terms should be clear; however, in the
event of any latent ambiguity, definitions provided herein take precedence over any dictionary or extrinsic definition.
As utilized in accordance with the present disclosure, the following terms, unless otherwise indicated, shall be understood to have the following meanings.
The term "BAX/BCL-2 modulators" as used herein refers to the agents that can modulate the expression or activity of BAX/BCL-2 homodimer.
The term "alkyl" as used herein refers to a saturated straight-chain or branched hydrocarbon group having 1 to 6 carbon atoms, especially 1 to 4 carbon groups, for example methyl, ethyl, propyl, 1-methylethyl, butyl, 1-methylpropyl, 2-methylpropyl, 1, 1-dimethylethyl, pentyl, 1-methylbutyl, 2-methylbutyl, 3-methylbutyl, 2, 2-dimethylpropyl, 1-ethylpropyl, hexyl, 1, 1-dimethylpropyl, 1, 2-dimethylpropyl, 1-methylpentyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 1, 1-dimethylbutyl, 1, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2, 2-dimethylbutyl, 2, 3-dimethylbutyl, 3, 3-dimethylbutyl, 1-ethylbutyl, 2-ethylbutyl, 1, 1, 2-trimethylpropyl, 1, 2, 2-trimethylpropyl, 1-ethyl-1-methylpropyl, 1-ethyl-2-methylpropyl. C1-C4-alkyl means for example methyl, ethyl, propyl, 1-methylethyl, butyl, 1-methylpropyl, 2-methylpropyl or 1, 1-dimethylethyl.
The term "heterocyclyl" as used herein refers to a monocyclic radical having 5 to 8 ring members, wherein in each case 1, 2, 3 or 4 of these ring members are heteroatoms selected, independently from each other, from the group consisting of oxygen, nitrogen and sulfur.
The term "preventing" or "prevention" as used herein refers to delaying the onset of the symptoms of a susceptible subject or reducing the occurrence of a disease.
The term "treating" or "treatment" as used herein denotes reducing and/or improving the symptoms of a susceptible subject or increasing the survival rate of the subject with certain lethal disorders or conditions.
The term "BAX/BCL-2-related disorders or conditions" as used herein denotes the disorders or conditions wherein the modulation of BAX/BCL-2 is beneficial. For example, such disorders or conditions include cancers, myeloproliferative disorder, prostate epithelial dysplasia, lymphangioleiomyomatosis, Kimura disease, and keloid.
The term "subject" as used herein denotes animals, especially mammals. In one preferred embodiment, the term "subject" denotes humans. In another preferred embodiment, the term "subject" denotes companion animals, such as cats and dogs.
The term "therapeutically effective amount" as used herein refers to the amount of an active ingredient used alone or in combination with other treatments/medicaments for treating PPARγ-related disorders or conditions that shows therapeutic efficacy.
The term "carrier" or "pharmaceutically acceptable carrier" refers to particles that can encapsulate active pharmaceutical ingredients. Examples of carriers suitable for the present invention include niosomes, polymersomes, nanoparticles, liposomes, nano suspended particles, solid lipid nanoparticles, magnetic nano-carriers, micelles, macromolecular conjugates, particulate drug carriers, and the like.
Unless otherwise required by context, singular terms shall include the plural and plural terms shall include the singular.
The inventors of the invention surprisingly found that compounds having a pyrimido [5, 4-d] pyrimidine structure can enhance the expression and activity of BAX/BCL-2 homodimer, and thus may serve as novel types of BAX/BCL-2 modulators. In a preferred embodiment, the pyrimido [5, 4-d] pyrimidine compound is dipyridamole.
The present invention thus provides a method of preventing or treating BAX/BCL-2-related disorders or conditions, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of formula (I) :
wherein each of R1, R2, R3 and R4 is independently selected from the group consisting of heterocyclyl and di (hydroxyalkyl) amino,
or a pharmaceutically acceptable salt thereof.
In an embodiment, R1 and R3 are heterocyclyl, preferably piperidyl, and R2 and R4 are di(hydroxyalkyl) amino, preferably N, N-di (hydroxyethyl) amino.
In a preferred embodiment, the compound is dipyridamole.
In another embodiment, the compound is encapsulated in a carrier, such as a niosome, a polymersome, a nanoparticle, a liposome, a nano suspended particle, a solid lipid nanoparticle, a magnetic nano-carrier, a micelle, a macromolecular conjugate or a particulate drug carrier.
In a preferred embodiment, the carrier is a liposome. In another embodiment, the liposome has a diameter in the range of about 50-700 nm, preferably about 60-530, more preferably 80-350 nm, and most preferably about 130-230 nm.
It is known in the art that when dipyridamole is administered in free form, it binds to the ENT receptors on a cell membrane and activates signaling pathways that block adenosine from passing into the cell. The inventors found that dipyridamole can regulate the expression of apoptosis/anti-apoptosis proteins and cause cell apoptosis through cell-penetrating drug delivery systems into the cell. Activation of the apoptosis pathway of cancer cells can facilitate the treatment of cancers known to be associated with abnormal apoptosis/anti-apoptosis protein expression.
Figure 1a shows that if dipyridamole is administered in free form, it mainly acts outside of cells and will promote the accumulation of adenosine, which will lead to reduced apoptosis. Figure 1b shows that dipyridamole inside the cell can activate the expression of apoptosis/anti-apoptosis proteins. In such case, the unfavorable action of dipyridamole outside the cell can be avoided. In the Examples below, the present invention demonstrates that by delivering dipyridamole directly into cells, the binding to the receptors on the cell membrane can be avoided so as to reduce the side effects such as oxidative stress and vasoconstriction caused by the accumulation of adenosine.
In an embodiment, the compound of formula (I) of the invention is encapsulated in a carrier for delivery into the cell. In a preferred embodiment, the carrier is a niosome, a polymersome, a nanoparticle, a liposome, a nano suspended particle, a solid lipid nanoparticle, a magnetic nano-carrier, a micelle, a macromolecular conjugate or a particulate drug carrier. Preferably, the carrier is a liposome. The liposome suitable for the present invention has a diameter in the range of about 10-300 nm, preferably about 80-280 nm, more preferably about 120-270 nm.
In another embodiment of the invention, the carriers may be niosomes, polymersomes, or polymers that have a diameter of less than 1 μm. Modifications can be made based on surface electric potential, hydrophilicity/hydrophobicity, size, morphology, shape and/or surface curvature.
The liposome formulation of the invention may comprise vesicles of various natures (e.g., unilamellar or multilamellar) , compositions, sizes, and characteristics, enclosing an
aqueous medium of diverse compositions, pH and osmotic strength. In a preferred embodiment, the main constituents of the liposome lipid layer membrane are selected from the group consisting of natural or synthetic phospholipids such as those listed below:
-1, 2-Dilauroyl-sn-Glycero-3-Phosphocholine (DLPC)
-1, 2-Dimyristoyl-sn-Glycero-3-Phosphocholine (DMPC)
-1, 2-Dipalmitoyl-sn-Glycero-3-Phosphocholine (DPPC)
-1, 2-Distearoyl-sn-Glycero-3-Phosphocholine (DSPC)
-1, 2-Dioleoyl-sn-Glycero-3-Phosphocholine (DOPC)
-1, 2-Dimyristoyl-sn-Glycero-3-Phosphoelhanolamine (DMPE)
-1, 2-Dipalmitoyl-sn-Glycero-3-Phosphoelhanolamine (DPPE)
-1, 2-Distearoyl-sn-Glycero-3-Phosphoelhanolamine (DSPE)
-1, 2-Dioleoyl-sn-Glycero-3-Phosphoelhanolamine (DOPE)
-1-Myristoyl-2-Palmitoyl-sn-Glycero-3-Phosphocholine (MPPC)
-1-Palmitoyl-2-Myristoyl-sn-Glycero-3-Phosphocholine (PMPC)
-1-Stearoyl-2-Palmitoyl-sn-Glycero-3-Phosphocholine (SPPC)
-1-Palmitoyl-2-Stearoyl-sn-Glycero-3-Phosphocholine (PSPC)
-1, 2-Dimyristoyl-sn-Glycero-3- [Phospho-rac- (1-glycerol) ] (DMPG)
-1, 2-Dipalmitoyl-sn-Glycero-3- [Phospho-rac- (1-glycerol) ] (DPPG)
-1, 2-Distearoyl-s/i-Glycero-3- [Phospho-rac- (1-glycerol) ] (DSPG)
-1, 2-Dioleoyl-sn-Glycero-3- [Phospho-rac- (1-glycerol) ] (DOPG)
-1, 2-Dimyristoyl-sn-Glycero-3-Phosphate (DMPA)
-1, 2-Dipalmitoyl-sn-Glycero-3-Phosphate (DPPA)
-1, 2-Dipalmitoyl-sn-Glycero-3- [Phospho-L-Serine] (DPPS)
-phosphatidylserine (PS) , and
-Natural L-a-phosphatidylcholine (from chicken egg, EPC, or from soy, SPC and HSPC) .
Preferred phospholipids are long saturated phospholipids, e.g. those having alkyl chains of more than 12, preferably more than 14, more preferably more than 16, most preferably more than 18 carbon atoms.
Preferred liposome compositions for use according to the invention are preferably those in which the liposomes are uni-and/or multilamellar, and comprise:
(i) 1 to 100, preferably 40 to 70 mol%physiologically acceptable phospholipids, preferably selected from the group consisting of DLPC, DMPC, DPPC, DSPC, DOPC, DMPE, DPPE, DSPE, DOPE, MPPC, PMPC, SPPC, PSPC, DMPG, DPPG, DSPG, DOPG, DMPA, DPPA, DPPS, PS, EPC, SPC and HSPC;
(ii) 1 to 100, preferably 40 to 70 mol%sphingolipids, preferably sphingomyelin;
(iii) 1 to 100, preferably 40 to 70 mol%surfactants, preferably featuring hydrophobic alkyl ethers (e.g. Brij) , alkyl esters, polysorbates, sorbitan esters, and/or alkyl amides;
(iv) 5 to 100, preferably 50 to 100 mol%amphiphilic polymers and/or co-polymers, preferably block copolymers comprising at least one block of a hydrophilic polymer or copolymer such as polyethylene glycol, and at least one block of a hydrophobic polymer or copolymer such as poly (lactide) , poly (caprolactone) , poly (butylene oxide) , poly (styrene oxide) , poly (styrene) , poly (ethylethylene) , or polydimethylsiloxanes,
(v) 0 to 60 mol%, preferably 20 to 50 mol%toxin retention-enhancing compounds, preferably sterol derivatives, preferably cholesterol; or
(vi) 0 to 30 mol%, preferably 1 to 5 mol%steric stabilizers, preferably PEGylated compounds, preferably PEGylated lipids, more preferably DSPE-PEG.
In a preferred embodiment, liposome-like vesicles are made from polymers and comprise no lipids, for which reason they are formally not considered liposomes but are called polymersomes. However, for the purpose of the present invention, polymersomes are meant to be encompassed by the term liposome as used for defining the invention and the claims.
Similarly, liposome-like vesicles made from synthetic surfactants and comprising no lipids are called niosomes. However, for the purpose of the present invention, niosomes are meant to be encompassed by the term liposome as used for defining the invention and the claims.
In an embodiment of the invention, polymerization of different high molecular polymers can be used, which comprise those in tri-block copolymer form such as ABA and BAB, and those in block copolymer form such as PLLA-PEG, PLGA-PEG, PLA-PEG, PLLA-mPEG, PLGA-mPEG and PLA-mPEG. Various shapes such as asterisk and L form can be designed, including block copolymers of PEG- (PLGA) 8, PEG- (PLLA) 8 and PEG- (PDLA) 8 Star. PEGylated modification can be used to modify any vehicle such as polymeric vehicle and liposome to achieve the effect of reducing the binding rate of plasma proteins (see Park, J. et al., (2009) "PEGylated PLGA nanoparticles for the improved delivery of doxorubicin.
Nanomedicine. " 5 (4) : 410-418. ; Lück, M. et al., (1998) "Plasma protein adsorption on biodegradable microspheres consisting of poly (D, L-lactide-co-glycolide) , poly (L-lactide) or ABA triblock copolymers containing poly (oxyethylene) . Influence of production method and polymer composition. " J. Control Release. 55 (2-3) : 107-20. ; and Sempf, K. et al, (2013) "Adsorption of plasma proteins on uncoated PLGA nanoparticles. " Eur. J. Pharm. Biopharm. 85 (1) : 53-60) .
The animal dose should not be extrapolated to a human equivalent dose (HED) by a simple conversion based on body weight. The Food and Drug Administration has suggested that the extrapolation of animal dose to human dose is correctly performed only through normalization to BSA, which often is represented in mg/m2. The human dose equivalent can be more appropriately calculated by using the formula: HED (mg/kg) = Animal dose (mg/kg) multiplied by Animal Km/Human Km. To convert the dose used in a mouse to a dose based on surface area for humans, multiply 22.4 mg/kg (Baur’s mouse dose) by the Km factor (3) for a mouse and then divide by the Km factor (37) for a human (see Table below) .
Values based on data from FDA Draft Guidelines
To convert a dose expressed in mg/kg to dose in mg/m2, multiply by Km value. According to the present invention, the effective dose of liposome-dipyridamole in mice is 10 mg/kg-100 mg/kg, in hamster 6-60 mg/kg, in rat 5-50 mg/kg, in guinea pig 3.75-37.5 mg/kg, in rabbit 2.5-25 mg/kg, in monkey 2.5-25 mg/kg, in dog 1.5-15 mg/kg, in cat 2.4-24 mg/kg, in baboon 1.5-15 mg/kg, in child 1.2-12 mg/kg, and in adult 0.81-8.1 mg/kg. Taking into consideration the differences in drug sensitivity among species, the broadest dose range without
limiting the species is 0.4-160 mg/kg, preferably 0.6-120 mg/kg, more preferably 0.8 mg/kg-100 mg/kg.
In an embodiment, the dipyridamole liposome may be used for treating cancer. In an embodiment, the cancers are drug-resistant or non-drug-resistant. In an embodiment, the cancers are breast cancers and liver cancers.
In an embodiment, the dipyridamole liposome may be administered in combination with other anti-cancer drugs, such as alkylating agents, antimetabolites, antibiotics, hormones, immune modulators, mitotic inhibitors, target therapeutic agents, and platinum drugs. In an embodiment, the anti-cancer drugs comprise chemotherapeutic drugs and target drugs. In an embodiment, the chemotherapeutic drugs comprise alkylating agents (such as Cyclophosphamide, Mechlorethamine and Melphalan ) , antimitotic agents (Vinblastine, Vincristine and Taxol) , DNA intercalating agents (Daunorubicin and Doxorubicin) , DNA cleaving agents, antimetabolites agents (such as Capecitabine, Cladribine, Cytarabine, Fludarabine phosphate, 5-Fluorouracil, Gemcitabine, 6-Mercaptopurine, Methotrexate (Amethopterin,MTX) , Mitoxantron, Pemetrexed disodium (Heptahydrate) , and Tegafur (FT-207) ) and topoisomerase inhibitors (such as (Camptothecin, Topotecan, Irinotecan, Podophyllotoxin and Etoposide) . In an embodiment, the target drugs comprise Trastuzumab, Lapatinib, Genfitinib, Erlotinib, Cetuximab, Bevacizumab, Rituximab, Bortizomib, Imatinib, Sunitinib, Rapamycin, Sirolimus, Temsirolimus, Everolimus, Ridaforolimus (deforolimus) and Sorafenib. In an embodiment, the other anti-cancer drug is 5-Fluorouracil (5-FU) .
Having now generally described the invention, the same may be more readily understood through reference to the following examples, which provide exemplary protocols for the production of the pharmaceutical composition of the invention and its use in the treatment of cancers. The examples are offered for illustrative purposes only, and are not intended to limit the scope of the present invention in any way. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperatures, etc. ) , but some experimental error and deviation should, of course, be allowed for.
Examples
Example 1: Preparation of dipyridamole liposome
Liposomes were prepared with positive and neutral charge containing phospholipid and cholesterol, wherein the mole percent of cholesterol was 5%to 75%. Small unilamellar vesicles were prepared. The dried lipid films were hydrated with an ammonium sulfate and sequentially extruded through a series of polycarbonate membrane filters. Dipyridamole was encapsulated into the liposomes via a transmembrane pH gradient or dehydration-rehydration, and the diameters of the extruded liposomes were in the range of 50-602 nm (dissolved in glucose solution) via the above manufacturing processes. The diameter of the liposome-dipyridamole was about 50 to 602 nm as shown in Table 1.
Table 1
Example 2: Cell viability (MTT assay) and Western blot analysis of the BCL-2 protein family
Cancer cell line culture
The medium for human breast adenocarcinoma is L-15 Medium. To make the complete growth medium, the following components were added to the base medium: fetal bovine serum to a final concentration of 10%. The cultures were incubated at 37℃ without CO2. The tested cancer cell lines include MDA-MB-231, PANC-1, BXPC-3, HCT-116, HT29, A375, and MeWo.
A stock solution of MTT (3- (4, 5-dimethylthiazol-2-yl) -2, 5-diphenyl-tetrazolium bromide, Sigma, St. Louis) was prepared by dissolving 5 mg MTT/ml in phosphate-buffered saline pH 7.5 and filtered through a 0.22 μm filter. The MTT assay used was essentially similar to that originally described by Mosmann (1983) . Briefly, 100μL of MTT solution (0.5 mg/mL) was added to the monolayers of the treated cancer cell lines and the microplates were incubated at 37℃ for 3h. 100μL of DMSO was added to each well and mixed thoroughly to dissolve the
dark blue crystals. The plates were read on a microELISA reader using a test wavelength of 570 nm and reference wavelength of 630 nm.
After the treatment, the cells were washed with 150mL RIPA cell lysis buffer (10mM Hepes pH=7.9, 1.5mM MgCl2, 10mM KCl, 1.0mM DTT, 0.1% Triton-X 100) , and centrifuged at 1200g for 10 minutes at 4℃. Supernatant containing cancer cell total proteins was then collected and analyzed by Western blotting. The Western blot method was performed as follows: protein concentration was measured using Bradford assay; 6X sample buffer (0.8 mM Tris-HCl, 10 mM EDTA, 10%SDS, 60%glycerol, 0.6 M β-mercaptoethano, 0.06%bromophenol blue, pH 6.8) was added into 50 μg of whole cell proteins; and an equal volume of lysis buffer was added to the samples. After being heated at 95℃ for 10 minutes to denature the proteins, the samples were immediately cooled on ice.
The samples were then separated by 12%SDS-PAGE electrophoresis (100 V) and transferred from the SDS-PAGE gels to PVDF membranes by wet blotting. The PVDF membranes were then treated with 5%skimmed milk at room temperature for 60 minutes to block non-specific binding. The membranes were incubated with primary antibody overnight at 4℃ and washed three times with PBST. The membranes were incubated with a secondary antibody at room temperature for 60 minutes and washed three times with PBST. The membranes were then washed one more time with PBS and incubated with an enhanced chemiluminescence (ECL) substrate for detection. Photos of the images were taken using automated chemiluminescence and fluorescence imaging system (UVP Biospectrum) .
Example 2.1: Cell viability and Bax/BCL-2 ratio of MCF-7 in cells treated with the dipyridamole liposome
The cell line used in the assay was human breast adenocarcinoma cells, MCF-7. The cells were treated with the dipyridamole liposome (10 and 20 μM) or dipyridamole freeform (10 and 20 μM) . The results are shown in Figures 4a and 4b, and normalized in Figures 5a and 5b and Table 2.
Table 2
Example 3: Cell viability and Bax/BCL-2 ratio of MDA-MB-231 in cells treated with the dipyridamole liposome in combination with Rapamycin
The cell line used in the assay was human breast adenocarcinoma cells, MDA-MB-231. The cells were treated with rapamycin (100 μM) , the dipyridamole liposome (3.125 and 25 μM) or dipyridamole freeform (3.125 and 25 μM) in combination with rapamycin (100 μM) . The results are shown in Table 3.
Table 3
Example 4: Cell viability and Bax/BCL-2 ratio in PANC-1 cells treated with the dipyridamole liposome in combination with Rapamycin
The cell line used in the assay was human pancreas adenocarcinoma cells, PANC-1. The cells were treated with rapamycin (10 μM) , the dipyridamole liposome (3.125 and 25 μM) or dipyridamole freeform (3.125 and 25 μM) in combination with rapamycin (10 μM) . The results are shown in Table 4.
Table 4
Example 5: Cell viability and Bax/BCL-2 ratio in PANC-1, BxPC-3, HCT-116, HT29, A375 and MeWo cells treated with the dipyridamole liposome in combination with 5-Fu
The cells were treated with 5-Fu (10 μM and 100 μM) , the dipyridamole liposome (3.125 and 25 μM) or dipyridamole freeform (25 μM) in combination with 5-Fu (10 μM) . The results are shown in Table 5.
Table 5
The results in this example demonstrate that by using dipyridamole liposome in combination with 5-Fu, the cytotoxic effect of these drugs on pancreatic, liver, breast and skin cancer cells is increased through balancing Bcl-2 and Bax ratio. The doses of these drugs may also be reduced. For example, the dose of 5-Fu may be reduced by at least 8 fold.
Further, through statistical analysis, it is found that among the 8 cancer cell lines tested, the increase of Bax/Bcl-2 ratio by dipyridamole liposome treatment negatively correlates to cancer cell survival rate (R=-0.789, p < 0.01) . This result proves that dipyridamole produces a synergistic effect with 5-Fu in inhibiting the growth of various types of cancer cell lines. Furthermore, a pharmaceutical composition comprising dipyridamole and 5-Fu can enhance the cytotoxic effect of 5-Fu in cancer cells, especially in cancers that are resistant to 5-Fu treatment (Fig. 2) .
Example 6: Cell viability of A375 and MeWo cells treated with the dipyridamole liposome in combination with Gemcitabine
In A375 and MeWo cancer cell lines, it is found that dipyridamole liposome exhibits a membrane-penetrating effect. By this membrane-penetrating effect, dipyridamole reduces antagonism to Gemcitabine as effective by 20-100%. This result proves that dipyridamole liposome increases penetration of drugs into the cells. The results are shown in Table 6. Extracellular dipyridamole blocks ENT-1 and prevents Gemcitabine from entering into
the cancer cells to produce its cytotoxic effect. The results in this example demonstrate that dipyridamole liposome reduces this phenomenon by 20-40%, indicating that dipyridamole liposome enhances the ratio of drugs that enter the cells.
Table 6
Other chemotherapeutic drugs, for example, the rapamycin analogs (Rapalogs) shown in Figure 3, may be used in combination with dipyridamole liposome of the present invention.
From the data above, it is found that dipyridamole liposome of the present invention produces a synergistic effect in inhibiting cancer cell proliferation when administered together with a chemotherapeutic drug by 10-40%more cell death (Fig. 4) . The synergistic effect in cancer cell death may result not simply from increasing Bax or decreasing Bcl-2, but increasing the Bax/Bcl-2 ratio (Figs. 5 and 6) . According to the examples, dipyridamole increases Bax/Bcl-2 ratio by 25-200%after entering into the cells, and increases inhibition on cancer cell proliferation by 20-50%. When combined with 5-Fu, dipyridamole inhibits cancer cell proliferation by 5-50%through entering into the cells and increasing Bax/Bcl-2 ratio by 15-50%. Combination of dipyridamole with rapamycin inhibits cancer cell proliferation by 10-30%through entering into the cells and increasing Bax/Bcl-2 ratio by 10-200%.
Numerous modifications and variations of the invention as set forth in the above illustrative examples are expected to occur to those skilled in the art. Consequently, only such limitations as appear in the appended claims should be placed on the invention.
Claims (20)
- A method of preventing or treating BAX/BCL-2-related disorders or conditions, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of formula (I) :wherein each of R1, R2, R3 and R4 is independently selected from the group consisting of heterocyclyl and di (hydroxyalkyl) amino,or a pharmaceutically acceptable salt thereof.
- The method of Claim 1, wherein R1 and R3 are heterocyclyl and R2 and R4 are di (hydroxyalkyl) amino.
- The method of Claim 1, wherein the heterocyclyl is piperidyl.
- The method of Claim 1, wherein the di (hydroxyalkyl) amino is N, N-di (hydroxyethyl) amino.
- The method of Claim 1, wherein the compound is dipyridamole.
- The method of Claim 1, wherein the compound is encapsulated in a carrier.
- The method of Claim 6, wherein the carrier is a niosome, a polymersome, a nanoparticle, a liposome, a nano suspended particle, a solid lipid nanoparticle, a magnetic nano-carrier, a micelle, a macromolecular conjugate or a particulate drug carrier.
- The method of Claim 7, wherein the carrier is a liposome.
- The method of Claim 8, wherein the liposome has a diameter in the range of about 50-602 nm.
- The method of Claim 1, wherein the subject is a human or non-human mammal.
- The method of Claim 10, wherein the non-human mammal is a cat or a dog.
- The method of Claim 1, wherein the BAX/BCL-2-related disorders or conditions are cancers, myeloproliferative disorder, prostate epithelial dysplasia, lymphangioleiomyomatosis, Kimura disease, and keloid.
- The method of Claim 12, wherein the cancer is drug-resistant or non-drug-resistant cancer.
- The method of Claim 13, wherein the cancer is breast cancer, liver cancer, gastric cancer, pancreatic cancer, kidney cancer, colorectal cancer, or lung cancer.
- The method of Claim 1, wherein the compound of formula (I) is administered alone or in combination with other anti-cancer drugs.
- The method of Claim 15, wherein the other anti-cancer drugs comprise chemotherapeutic drugs and target drugs.
- The method of Claim 16, wherein the chemotherapeutic drugs comprise alkylating agents, antimitotic agents, DNA intercalating agents, DNA cleaving agents, antimetabolites agents and topoisomerase inhibitors.
- The method of Claim 17, wherein the chemotherapeutic drugs comprise Cyclophosphamide, Mechlorethamine, Melphalan, Vinblastine, Vincristine, Taxol, Daunorubicin, Doxorubicin, Cytarabine, 5-Fluorouracil, 6-Mercaptopurine, Methotrexate, Camptothecin, Topotecan, Irinotecan, Podophyllotoxin and Etoposide.
- The method of Claim18, wherein the chemotherapeutic drug is 5-Fluorouracil.
- The method of Claim 16, wherein the target drugs comprise Trastuzumab, Lapatinib, Genfitinib, Erlotinib, Cetuximab, Bevacizumab, Rituximab, Bortizomib, Imatinib, Sunitinib, Rapamycin, Sirolimus, Temsirolimus, Everolimus, Ridaforolimus (deforolimus) and Sorafenib.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US201662367850P | 2016-07-28 | 2016-07-28 | |
| US62/367,850 | 2016-07-28 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2018019284A1 true WO2018019284A1 (en) | 2018-02-01 |
Family
ID=61015522
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/CN2017/094855 Ceased WO2018019284A1 (en) | 2016-07-28 | 2017-07-28 | Compounds for enhancing bax/bcl-2 expression and activity and therapeutic use thereof |
Country Status (2)
| Country | Link |
|---|---|
| TW (1) | TWI721202B (en) |
| WO (1) | WO2018019284A1 (en) |
Cited By (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2021002986A3 (en) * | 2019-05-31 | 2021-05-27 | Case Western Reserve University | Bax inhibitors and uses thereof |
| CN116173035A (en) * | 2021-11-26 | 2023-05-30 | 中国科学院深圳先进技术研究院 | Application of dipyridamole in inhibiting Mcl-1 |
| WO2023092462A1 (en) * | 2021-11-26 | 2023-06-01 | 中国科学院深圳先进技术研究院 | Use of dipyridamole in inhibiting mcl-1 |
Citations (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN1768754A (en) * | 2004-11-05 | 2006-05-10 | 刘国廉 | Application of dipyridamole in preparation of anti-malignant tumor medicine |
Family Cites Families (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2012097133A2 (en) * | 2011-01-12 | 2012-07-19 | H. Lee Moffitt Cancer Center And Research Institute, Inc. | Compounds and methods for inducing apoptosis in cancer cells using a bh3 alpha-helical mimetic |
-
2017
- 2017-07-28 WO PCT/CN2017/094855 patent/WO2018019284A1/en not_active Ceased
- 2017-07-28 TW TW106125621A patent/TWI721202B/en active
Patent Citations (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN1768754A (en) * | 2004-11-05 | 2006-05-10 | 刘国廉 | Application of dipyridamole in preparation of anti-malignant tumor medicine |
Cited By (7)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2021002986A3 (en) * | 2019-05-31 | 2021-05-27 | Case Western Reserve University | Bax inhibitors and uses thereof |
| JP2022534902A (en) * | 2019-05-31 | 2022-08-04 | ケース ウエスタン リザーブ ユニバーシティ | BAX inhibitors and uses thereof |
| JP2025066800A (en) * | 2019-05-31 | 2025-04-23 | ケース ウエスタン リザーブ ユニバーシティ | Bax inhibitors and uses thereof |
| JP7675020B2 (en) | 2019-05-31 | 2025-05-12 | ケース ウエスタン リザーブ ユニバーシティ | BAX INHIBITORS AND USES THEREOF |
| US12479857B2 (en) | 2019-05-31 | 2025-11-25 | Case Western Reserve University | Bax inhibitors and uses thereof |
| CN116173035A (en) * | 2021-11-26 | 2023-05-30 | 中国科学院深圳先进技术研究院 | Application of dipyridamole in inhibiting Mcl-1 |
| WO2023092462A1 (en) * | 2021-11-26 | 2023-06-01 | 中国科学院深圳先进技术研究院 | Use of dipyridamole in inhibiting mcl-1 |
Also Published As
| Publication number | Publication date |
|---|---|
| TWI721202B (en) | 2021-03-11 |
| TW201805001A (en) | 2018-02-16 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| Gajbhiye et al. | Lipid polymer hybrid nanoparticles: a custom-tailored next-generation approach for cancer therapeutics | |
| Kong et al. | Current status of sorafenib nanoparticle delivery systems in the treatment of hepatocellular carcinoma | |
| Allahou et al. | Investigating the application of liposomes as drug delivery systems for the diagnosis and treatment of cancer | |
| Palazzolo et al. | The clinical translation of organic nanomaterials for cancer therapy: a focus on polymeric nanoparticles, micelles, liposomes and exosomes | |
| KR102584446B1 (en) | Alpha and gamma-D polyglutamate antifolates and uses thereof | |
| Haggag et al. | Co-delivery of a RanGTP inhibitory peptide and doxorubicin using dual-loaded liposomal carriers to combat chemotherapeutic resistance in breast cancer cells | |
| Boulikas | Molecular mechanisms of cisplatin and its liposomally encapsulated form, Lipoplatin™. Lipoplatin™ as a chemotherapy and antiangiogenesis drug | |
| US11090367B2 (en) | Restoration of tumor suppression using mRNA-based delivery system | |
| Zeng et al. | Application of functional vincristine plus dasatinib liposomes to deletion of vasculogenic mimicry channels in triple-negative breast cancer | |
| Fatfat et al. | Micelles as potential drug delivery systems for colorectal cancer treatment | |
| US20180110831A1 (en) | Cd 47 containing porous nanoparticle supported lipid bilayers (protocells) field of the invention | |
| Qi et al. | Research progress of SN38 drug delivery system in cancer treatment | |
| JP2016529284A (en) | Nanoparticle drug delivery system and method for treating cancer and neurotrauma | |
| WO2013173693A1 (en) | Nanoparticles with enhanced entry into cancer cells | |
| Desai et al. | Pancreatic cancer: Recent advances in nanoformulation-based therapies | |
| Du et al. | F7 and topotecan co-loaded thermosensitive liposome as a nano-drug delivery system for tumor hyperthermia | |
| Cano et al. | Recent advances on antitumor agents-loaded polymeric and lipid-based nanocarriers for the treatment of brain cancer | |
| CN102357074B (en) | Anti-tumor multi-medicine resistant targeted liposome | |
| WO2008038291A1 (en) | Combination of liposomal anti-cancer drugs and lysosome/endosome ph increasing agents for therapy | |
| Sa et al. | Responsive role of nanomedicine in the tumor microenvironment and cancer drug resistance | |
| Li et al. | Thermo-Sensitive Liposome co-Loaded of Vincristine and Doxorubicin Based on Their Similar Physicochemical Properties had Synergism on Tumor Treatment. | |
| TW202207904A (en) | Liposome formulations for treatment of cancers and drug resistance of cancers | |
| WO2018019284A1 (en) | Compounds for enhancing bax/bcl-2 expression and activity and therapeutic use thereof | |
| TWI725008B (en) | COMPOUNDS FOR ENHANCING PPARγ EXPRESSION AND NUCLEAR TRANSLOCATION AND THERAPEUTIC USE THEREOF | |
| Mukhopadhyay | Nano drugs: A critical review of their patents and market |
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: 17833587 Country of ref document: EP Kind code of ref document: A1 |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| 32PN | Ep: public notification in the ep bulletin as address of the adressee cannot be established |
Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205N DATED 05-04-2019) |
|
| 122 | Ep: pct application non-entry in european phase |
Ref document number: 17833587 Country of ref document: EP Kind code of ref document: A1 |