1
|
Haque MA, Shrestha A, Mikelis CM, Mattheolabakis G. Comprehensive analysis of lipid nanoparticle formulation and preparation for RNA delivery. Int J Pharm X 2024; 8:100283. [PMID: 39309631 PMCID: PMC11415597 DOI: 10.1016/j.ijpx.2024.100283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/21/2024] [Accepted: 09/07/2024] [Indexed: 09/25/2024] Open
Abstract
Nucleic acid-based therapeutics are a common approach that is increasingly popular for a wide spectrum of diseases. Lipid nanoparticles (LNPs) are promising delivery carriers that provide RNA stability, with strong transfection efficiency, favorable and tailorable pharmacokinetics, limited toxicity, and established translatability. In this review article, we describe the lipid-based delivery systems, focusing on lipid nanoparticles, the need of their use, provide a comprehensive analysis of each component, and highlight the advantages and disadvantages of the existing manufacturing processes. We further summarize the ongoing and completed clinical trials utilizing LNPs, indicating important aspects/questions worth of investigation, and analyze the future perspectives of this significant and promising therapeutic approach.
Collapse
Affiliation(s)
- Md. Anamul Haque
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Archana Shrestha
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Constantinos M. Mikelis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras 26504, Greece
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - George Mattheolabakis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| |
Collapse
|
2
|
Lahooti B, Akwii RG, Patel D, ShahbaziNia S, Lamprou M, Madadi M, Abbruscato TJ, Astrinidis A, Bickel U, Al-Ahmad A, German NA, Mattheolabakis G, Mikelis CM. Endothelial-Specific Targeting of RhoA Signaling via CD31 Antibody-Conjugated Nanoparticles. J Pharmacol Exp Ther 2023; 385:35-49. [PMID: 36746610 PMCID: PMC10029826 DOI: 10.1124/jpet.122.001384] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 01/04/2023] [Accepted: 01/17/2023] [Indexed: 02/08/2023] Open
Abstract
Existing vascular endothelial growth factor-oriented antiangiogenic approaches are known for their high potency. However, significant side effects associated with their use drive the need for novel antiangiogenic strategies. The small GTPase RhoA is an established regulator of actin cytoskeletal dynamics. Previous studies have highlighted the impact of endothelial RhoA pathway on angiogenesis. Rho-associate kinase (ROCK), a direct RhoA effector, is potently inhibited by Fasudil, a clinically relevant ROCK inhibitor. Here, we aimed to target the RhoA signaling in endothelial cells by generating Fasudil-encapsulated CD31-targeting liposomes as a potential antiangiogenic therapy. The liposomes presented desirable characteristics, preferential binding to CD31-expressing HEK293T cells and to endothelial cells, inhibited stress fiber formation and cytoskeletal-related morphometric parameters, and inhibited in vitro angiogenic functions. Overall, this work shows that the nanodelivery-mediated endothelial targeting of RhoA signaling can offer a promising strategy for angiogenesis inhibition in vascular-related diseases. SIGNIFICANCE STATEMENT: Systemic administration of antiangiogenic therapeutics induces side effects to non-targeted tissues. This study, among others, has shown the impact of the RhoA signaling in the endothelial cells and their angiogenic functions. Here, to minimize potential toxicity, this study generated CD31-targeting liposomes with encapsulated Fasudil, a clinically relevant Rho kinase inhibitor, and successfully targeted endothelial cells. In this proof-of-principle study, the efficient Fasudil delivery, its impact on the endothelial signaling, morphometric alterations, and angiogenic functions verify the benefits of site-targeted antiangiogenic therapy.
Collapse
Affiliation(s)
- Behnaz Lahooti
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Racheal G Akwii
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Dhavalkumar Patel
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Siavash ShahbaziNia
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Margarita Lamprou
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Mahboubeh Madadi
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Thomas J Abbruscato
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Aristotelis Astrinidis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Ulrich Bickel
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Abraham Al-Ahmad
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Nadezhda A German
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - George Mattheolabakis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Constantinos M Mikelis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| |
Collapse
|
3
|
Long-Circulating and Fusogenic Liposomes Loaded with Paclitaxel and Doxorubicin: Effect of Excipient, Freezing, and Freeze-Drying on Quality Attributes. Pharmaceutics 2022; 15:pharmaceutics15010086. [PMID: 36678715 PMCID: PMC9866235 DOI: 10.3390/pharmaceutics15010086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 12/30/2022] Open
Abstract
Liposomes can increase plasma half-life, enhance targeting, and diminish the side-effects of loaded drugs. On the downside, physical and chemical instabilities of dispersions often result in a reduced lifespan, which limits their availability on the market. Solid formulations obtained by freeze-drying can immobilize vesicles and provide extended shelf life. For both processes, the choice of excipients and process parameters are crucial to protect the carrier layers against tension caused by freezing and/or dehydration. The aim of this work is to evaluate the influence of freezing and drying parameters, besides excipient choice, to obtain solid long-circulating and fusogenic liposomes (LCFL-PTX/DXR) co-encapsulating paclitaxel (PTX) and doxorubicin (DXR) at a synergistic ratio (1:10). METHODS LCFL-PTX/DXR was evaluated by freeze-drying microscopy (glass transition, Tg'), differential scanning calorimetry (collapse temperature, Tc), freeze-thawing and freeze-drying processes. Freeze-dried samples were evaluated by thermogravimetry (residual moisture) and the resuspended liposomes were characterized in terms of size, polydispersity index (PI), zeta potential (ZP), and drug content. Liposomes morphology was evaluated by cryomicroscopy. RESULTS Trehalose protected PTX cargo upon freeze-thawing and more than 80% of the original DXR retention. The formulations with trehalose resulted in a cake with 5-7% of moisture content (200-240 nm); 44-60% of PTX retention, and 25-35% of DXR retention, with the variations caused by cryoprotector concentration and process changes. CONCLUSIONS Trehalose protected liposome integrity, maintaining PTX retention and most of DXR upon freeze-thawing. Freeze-drying reduced the retention of both drugs inside all liposomes, whereas formulation with trehalose presented minor losses. Therefore, this frozen formulation is an alternative product option, with no need for manipulation before use.
Collapse
|
4
|
Alghurabi H, Tagami T, Ogawa K, Ozeki T. Preparation, Characterization and In Vitro Evaluation of Eudragit S100-Coated Bile Salt-Containing Liposomes for Oral Colonic Delivery of Budesonide. Polymers (Basel) 2022; 14:2693. [PMID: 35808738 PMCID: PMC9268925 DOI: 10.3390/polym14132693] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/15/2022] [Accepted: 06/15/2022] [Indexed: 12/13/2022] Open
Abstract
The aim of this study was to prepare a liposomal formulation of a model drug (budesonide) for colonic delivery by incorporating a bile salt (sodium glycocholate, SGC) into liposomes followed by coating with a pH-responsive polymer (Eudragit S100, ES100). The role of the SGC is to protect the liposome from the emulsifying effect of physiological bile salts, while that of ES100 is to protect the liposomes from regions of high acidity and enzymatic activity in the stomach and small intestine. Vesicles containing SGC were prepared by two preparation methods (sonication and extrusion), and then coated by ES100 (ES100-SGC-Lip). ES100-SGC-Lip showed a high entrapment efficiency (>90%) and a narrow size distribution (particle size = 275 nm, polydispersity index < 0.130). The characteristics of liposomes were highly influenced by the concentration of incorporated SGC. The lipid/polymer weight ratio, liposome charge, liposome addition, and mixing rate were critical factors for efficient and uniform coating. In vitro drug release studies in various simulated fluids indicate a pH-dependent dissolution of the coating layer, and the disintegration process of ES100-SGC-Lip was evaluated. In conclusion, the bile salt-containing ES100-coated liposomal formulation has potential for effective oral colonic drug delivery.
Collapse
Affiliation(s)
- Hamid Alghurabi
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan; (H.A.); (T.T.); (K.O.)
- Department of Pharmaceutics, College of Pharmacy, University of Kerbala, Kerbala 56001, Iraq
| | - Tatsuaki Tagami
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan; (H.A.); (T.T.); (K.O.)
| | - Koki Ogawa
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan; (H.A.); (T.T.); (K.O.)
| | - Tetsuya Ozeki
- Drug Delivery and Nano Pharmaceutics, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan; (H.A.); (T.T.); (K.O.)
| |
Collapse
|
5
|
Ramos-Inza S, Ruberte AC, Sanmartín C, Sharma AK, Plano D. NSAIDs: Old Acquaintance in the Pipeline for Cancer Treatment and Prevention─Structural Modulation, Mechanisms of Action, and Bright Future. J Med Chem 2021; 64:16380-16421. [PMID: 34784195 DOI: 10.1021/acs.jmedchem.1c01460] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The limitations of current chemotherapeutic drugs are still a major issue in cancer treatment. Thus, targeted multimodal therapeutic approaches need to be strategically developed to successfully control tumor growth and prevent metastatic burden. Inflammation has long been recognized as a hallmark of cancer and plays a key role in the tumorigenesis and progression of the disease. Several epidemiological, clinical, and preclinical studies have shown that traditional nonsteroidal anti-inflammatory drugs (NSAIDs) exhibit anticancer activities. This Perspective reports the most recent outcomes for the treatment and prevention of different types of cancers for several NSAIDs alone or in combination with current chemotherapeutic drugs. Furthermore, an extensive review of the most promising structural modifications is reported, such as phospho, H2S, and NO releasing-, selenium-, metal complex-, and natural product-NSAIDs, among others. We also provide a perspective about the new strategies used to obtain more efficient NSAID- or NSAID derivative- formulations for targeted delivery.
Collapse
Affiliation(s)
- Sandra Ramos-Inza
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, E-31008 Pamplona, Spain
| | - Ana Carolina Ruberte
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, E-31008 Pamplona, Spain
| | - Carmen Sanmartín
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, E-31008 Pamplona, Spain
| | - Arun K Sharma
- Department of Pharmacology, Penn State Cancer Institute, CH72, Penn State College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Daniel Plano
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, E-31008 Pamplona, Spain
| |
Collapse
|
6
|
Huang W, Wen Z, Saglam MS, Huang L, Honkanen RA, Rigas B. Phospho-Sulindac (OXT-328) Inhibits Dry Eye Disease in Rabbits: A Dose-, Formulation- and Structure-Dependent Effect. J Ocul Pharmacol Ther 2021; 37:321-330. [PMID: 34152861 DOI: 10.1089/jop.2019.0025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Purpose: Inflammation of the ocular surface is central to dry eye disease (DED). The anti-inflammatory agent phospho-sulindac (PS) at a high dose was efficacious against DED in a rabbit model. We assessed the dose, formulation and structure dependence of PS's effect. Methods: In rabbits with concanavalin A-induced DED we evaluated a range of PS concentrations (0.05%-1.6%) and dosing frequencies, assessed the duration of its effect with PS in 2 solution formulations and one emulsion formulation, and compared the efficacy of PS to that of sulindac, and of the structurally similar phospho-ibuprofen amide. We determined tear breakup time (TBUT) (tear stability), Schirmer's tear test (tear production), and by esthesiometry corneal sensitivity (symptoms). We also determined the biodistribution in the eye of topically applied PS. Results: PS in a solution formulation, given as eye drops q.i.d. was efficacious starting at a dose of 0.1%. The effect was apparent after 2 days of treatment and lasted at least 8 days after the last dose. Both signs (evidenced by TBUT and Schirmer's test) and symptoms (measured by corneal sensitivity) improved significantly. The best formulation was the solution formulation; a cyclodextrin-based formulation was also successful but the emulsion formulation was not. PS and its metabolites were essentially restricted to the anterior chamber of the eye. Sulindac and phospho-ibuprofen amide had no efficacy on DED. Conclusions: PS is efficacious against DED. Its effect, encompassing signs, and symptoms, are dose, formulation, and structure dependent. PS has therapeutic promise and merits further development.
Collapse
Affiliation(s)
- Wei Huang
- Department of Ophthalmology, Stony Brook University, Stony Brook, New York, USA.,Department of Medicine, Stony Brook University, Stony Brook, New York, USA.,Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziyi Wen
- Department of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Muhammet S Saglam
- Department of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Liqun Huang
- Department of Medicine, Stony Brook University, Stony Brook, New York, USA.,Medicon Pharmaceuticals, Setauket, New York, USA
| | - Robert A Honkanen
- Department of Ophthalmology, Stony Brook University, Stony Brook, New York, USA
| | - Basil Rigas
- Department of Preventive Medicine, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
7
|
Naziris N, Pippa N, Demetzos C. A Novel, Nontoxic and Scalable Process to Produce Lipidic Vehicles. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E5035. [PMID: 33171678 PMCID: PMC7664659 DOI: 10.3390/ma13215035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 10/28/2020] [Accepted: 11/05/2020] [Indexed: 12/26/2022]
Abstract
Lipidic vehicles are novel industrial products, utilized as components for pharmaceutical, cosmeceutical and nutraceutical formulations. The present study concerns a newly invented method to produce lipidic vehicles in the nanoscale that is simple, nontoxic, versatile, time-efficient, low-cost and easy to scale up. The process is a modification of the heating method (MHM) and comprises (i) providing a mixture of an amphiphilic lipid and a charged lipid and/or a fluidity regulator in a liquid medium composed of water and a liquid polyol, (ii) stirring and heating the mixture in two heating steps, wherein the temperature of the second step is higher than the temperature of the first step and (iii) allowing the mixture to cool down to room temperature. The process leads to the self-assembly of nanoparticles of small size and good homogeneity, compared with conventional approaches that require additional size reduction steps. In addition, the incorporation of bioactive molecules, such as drugs, inside the nanoparticles is possible, while lyophilization of the products provides long-term stability. Most importantly, the absence of toxic solvents and the simplicity guarantee the safety and scalability of the process, distinguishing it from most prior art processes to produce of lipidic vehicles.
Collapse
Affiliation(s)
| | | | - Costas Demetzos
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 15771 Athens, Greece; (N.N.); (N.P.)
| |
Collapse
|
8
|
Effect of Oleic Acid, Cholesterol, and Octadecylamine on Membrane Stability of Freeze-Dried Liposomes Encapsulating Natural Antimicrobials. FOOD BIOPROCESS TECH 2020. [DOI: 10.1007/s11947-020-02419-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
9
|
Gouda AM, Beshr EA, Almalki FA, Halawah HH, Taj BF, Alnafaei AF, Alharazi RS, Kazi WM, AlMatrafi MM. Arylpropionic acid-derived NSAIDs: New insights on derivatization, anticancer activity and potential mechanism of action. Bioorg Chem 2019; 92:103224. [PMID: 31491568 DOI: 10.1016/j.bioorg.2019.103224] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 08/08/2019] [Accepted: 08/26/2019] [Indexed: 12/28/2022]
Abstract
NSAIDs displayed chemopreventive and anticancer effects against several types of cancers. Moreover, combination of NSAIDs with anticancer agents resulted in enhanced anticancer activity. These findings have attracted much attention of researchers working in this field. The 2-arylpropionic acid-derived NSAIDs represent one of the most widely used anti-inflammatory agents. Additionally, they displayed antiproliferative activities against different types of cancer cells. Large volume of research was performed to identify molecular targets responsible for this activity. However, the exact mechanism underlying the anticancer activity of profens is still unclear. In this review article, the anticancer potential, structure activity relationship and synthesis of selected profen derivatives were summarized. This review is focused also on non-COX targets which can mediate the anticancer activity of this derivatives. The data in this review highlighted profens as promising lead compounds in future research to develop potent and safe anticancer agents.
Collapse
Affiliation(s)
- Ahmed M Gouda
- Department of Medicinal Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt.
| | - Eman A Beshr
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Faisal A Almalki
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Hadeel H Halawah
- B-Pharmacy Program, Faculty of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Batool Fawzi Taj
- B-Pharmacy Program, Faculty of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Athir Faiz Alnafaei
- B-Pharmacy Program, Faculty of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | | | - Weam Mahmood Kazi
- B-Pharmacy Program, Faculty of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Malak M AlMatrafi
- B-Pharmacy Program, Faculty of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| |
Collapse
|
10
|
Tang WL, Tang WH, Chen WC, Diako C, Ross CF, Li SD. Development of a Rapidly Dissolvable Oral Pediatric Formulation for Mefloquine Using Liposomes. Mol Pharm 2017; 14:1969-1979. [DOI: 10.1021/acs.molpharmaceut.7b00077] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Wei-Lun Tang
- Faculty
of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Wei-Hsin Tang
- Faculty
of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Weihsu Claire Chen
- Faculty
of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Charles Diako
- School
of Food Science, Washington State University, Pullman, Washington 99164, United States
| | - Carolyn F. Ross
- School
of Food Science, Washington State University, Pullman, Washington 99164, United States
| | - Shyh-Dar Li
- Faculty
of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| |
Collapse
|
11
|
Zhang T, Wang H, Ye Y, Zhang X, Wu B. Micellar emulsions composed of mPEG-PCL/MCT as novel nanocarriers for systemic delivery of genistein: a comparative study with micelles. Int J Nanomedicine 2015; 10:6175-84. [PMID: 26491290 PMCID: PMC4598212 DOI: 10.2147/ijn.s91348] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Polymeric micelles receive considerable attention as drug delivery vehicles, depending on the versatility in drug solubilization and targeting therapy. However, their use invariably suffers with poor stability both in in vitro and in vivo conditions. Here, we aimed to develop a novel nanocarrier (micellar emulsions, MEs) for a systemic delivery of genistein (Gen), a poorly soluble anticancer agent. Gen-loaded MEs (Gen-MEs) were prepared from methoxy poly(ethylene glycol)-block-(ε-caprolactone) and medium-chain triglycerides (MCT) by solvent-diffusion technique. Nanocarriers were characterized by dynamic light scattering, transmission electron microscopy, and in vitro release. The resulting Gen-MEs were approximately 46 nm in particle size with a narrow distribution. Gen-MEs produced a different in vitro release profile from the counterpart of Gen-ME. The incorporation of MCT significantly enhanced the stability of nanoparticles against dilution with simulated body fluid. Pharmacokinetic study revealed that MEs could notably extend the mean retention time of Gen, 1.57- and 7.38-fold as long as that of micelles and solution formulation, respectively, following intravenous injection. Furthermore, MEs markedly increased the elimination half-life (t1/2β) of Gen, which was 2.63-fold larger than that of Gen solution. Interestingly, Gen distribution in the liver and kidney for MEs group was significantly low relative to the micelle group in the first 2 hours, indicating less perfusion in such two tissues, which well accorded with the elongated mean retention time. Our findings suggested that MEs may be promising carriers as an alternative of micelles to systemically deliver poorly soluble drugs.
Collapse
Affiliation(s)
- Tianpeng Zhang
- Division of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou, People's Republic of China
| | - Huan Wang
- Division of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou, People's Republic of China
| | - Yanghuan Ye
- Division of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou, People's Republic of China
| | - Xingwang Zhang
- Division of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou, People's Republic of China
| | - Baojian Wu
- Division of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou, People's Republic of China
| |
Collapse
|
12
|
Choi JY, Ramasamy T, Tran TH, Ku SK, Shin BS, Choi HG, Yong CS, Kim JO. Systemic delivery of axitinib with nanohybrid liposomal nanoparticles inhibits hypoxic tumor growth. J Mater Chem B 2014; 3:408-416. [PMID: 32262043 DOI: 10.1039/c4tb01442a] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Axitinib (AXT) is a potent and selective orally administered inhibitor of the vascular endothelial growth factor receptors 1-3 that contribute to the pathogenesis of solid tumors. The goal of the present study was to enhance the antiangiogenic and antitumor effects of AXT under hypoxia. Here we developed spherical polypeptide-coated hybrid liposomal nanoparticles (P-LNP/AXT) with a narrow size distribution and high loading efficiency. The cytotoxic effects of P-LNP/AXT on cancer cells were lower than those of AXT, and the human cancer cell lines SCC7, BT-474, and SH-SY5YP efficiently incorporated P-LNP/AXT. However, these formulations were not significantly internalized by the mouse macrophage cell line RAW 264.7, suggesting that they could evade the reticuloendothelial system. Western blotting analysis showed a significant increase in the level of expression of hydroxy-HIF-1α when cells were treated with P-LNP/AXT. The growth of tumors in mice treated with P-LNP/AXT was significantly inhibited compared with controls. Further, elevated levels of caspase-3 and poly (ADP-ribose) polymerase and reduced levels of platelet/endothelial cell adhesion molecule 1 (PECAM1, CD31) and Ki-67 in tumor cells suggested that tumor cells underwent apoptosis and that angiogenesis was inhibited within the tumor. Thus, P-LNP/AXT shows promise for cancer chemotherapy by inhibiting tumor angiogenesis.
Collapse
Affiliation(s)
- Ju Yeon Choi
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan 712-749, Republic of Korea.
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Cheng KW, Nie T, Ouyang N, Alston N, Wong CC, Mattheolabakis G, Papayannis I, Huang L, Rigas B. A novel ibuprofen derivative with anti-lung cancer properties: synthesis, formulation, pharmacokinetic and efficacy studies. Int J Pharm 2014; 477:236-43. [PMID: 25311177 DOI: 10.1016/j.ijpharm.2014.10.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 07/22/2014] [Accepted: 10/07/2014] [Indexed: 12/16/2022]
Abstract
Phospho-non-steroidal anti-inflammatory drugs (phospho-NSAIDs) are a novel class of NSAID derivatives with potent antitumor activity. However, phospho-NSAIDs have limited stability in vivo due to their rapid hydrolysis by carboxylesterases at their carboxylic ester link. Here, we synthesized phospho-ibuprofen amide (PIA), a metabolically stable analog of phospho-ibuprofen, formulated it in nanocarriers, and evaluated its pharmacokinetics and anticancer efficacy in pre-clinical models of human lung cancer. PIA was 10-fold more potent than ibuprofen in suppressing the growth of human non-small-cell lung cancer (NSCLC) cell lines, an effect mediated by favorably altering cytokinetics and inducing oxidative stress. Pharmacokinetic studies in rats revealed that liposome-encapsulated PIA exhibited remarkable resistance to hydrolysis by carboxylesterases, remaining largely intact in the systemic circulation, and demonstrated selective distribution to the lungs. The antitumor activity of liposomal PIA was evaluated in a metastatic model of human NSCLC in mice. Liposomal PIA strongly inhibited lung tumorigenesis (>95%) and was significantly (p<0.05) more efficacious than ibuprofen. We observed a significant induction of urinary 8-iso-prostaglandin F2αin vivo, which indicates that ROS stress probably plays an important role in mediating the antitumor efficacy of PIA. Our findings suggest that liposomal PIA is a potent agent in the treatment of lung cancer and merits further evaluation.
Collapse
Affiliation(s)
- Ka-Wing Cheng
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Ting Nie
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Nengtai Ouyang
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Medicon Pharmaceuticals, Inc., Stony Brook, NY, USA
| | - Ninche Alston
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Chi C Wong
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Department of Medicine and Therapeutics, Chinese University of Hong Kong, Hong Kong
| | - George Mattheolabakis
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Ioannis Papayannis
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Liqun Huang
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Basil Rigas
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
14
|
Mattheolabakis G, Wong CC, Sun Y, Amella CA, Richards R, Constantinides PP, Rigas B. Pegylation improves the pharmacokinetics and bioavailability of small-molecule drugs hydrolyzable by esterases: a study of phospho-Ibuprofen. J Pharmacol Exp Ther 2014; 351:61-6. [PMID: 25047517 DOI: 10.1124/jpet.114.217208] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Esterase hydrolysis of drugs can accelerate their elimination, thereby limiting their efficacy. Polyethylene glycol (PEG) covalently attached to drugs (pegylation) is known to improve the efficiency of many drugs. Using as a test agent the novel phospho-ibuprofen (PI), we examined whether pegylation of PI could abrogate its hydrolytic degradation by esterases; PI, known to inhibit colon cancer growth, has a carboxylic ester hydrolyzable by carboxylesterases (CES). We covalently attached mPEG-2000 to PI (PI-PEG) and studied its stability by exposing it to cells overexpressing CES and by administering it to mice. We also evaluated PI-PEG's anticancer efficacy in human colon cancer xenografts and in Apc(min/+) mice. PI-PEG was stable in the presence of cells overexpressing CES1 or CES2, whereas PI was extensively hydrolyzed (90.2 ± 0.7%, 14.3 ± 1.1%, mean ± S.E.M.). In mice, PI was nearly completely hydrolyzed. Intravenous administration of PI-PEG resulted in significant levels in blood and in colon cancer xenografts (xenograft values in parentheses): area under the curve for 0-24 hours = 2351 (2621) (nmol/g) × h; Cmax = 1965 (886) nmol/g; Tmax = 0.08 (2) hour. The blood levels of ibuprofen, its main hydrolytic product, were minimal. Compared with controls, PI-PEG inhibited the growth of the xenografts by 74.8% (P < 0.01) and reduced intestinal tumor multiplicity in Apc(min/+) mice by 73.1% (P < 0.01), prolonging their survival (100% versus 55.1% of controls; P = 0.013). Pegylation protects PI from esterase hydrolysis and improves its pharmacokinetics. In preclinical models of colon cancer, PI-PEG is a safe and efficacious agent that merits further evaluation.
Collapse
Affiliation(s)
- George Mattheolabakis
- Department of Medicine, Stony Brook University, Stony Brook, New York (G.M., C.C.W., Y.S., C.A.A., R.R., B.R.); and Medicon Pharmaceuticals, Inc. (P.P.C., B.R.), Stony Brook, New York
| | - Chi C Wong
- Department of Medicine, Stony Brook University, Stony Brook, New York (G.M., C.C.W., Y.S., C.A.A., R.R., B.R.); and Medicon Pharmaceuticals, Inc. (P.P.C., B.R.), Stony Brook, New York
| | - Yu Sun
- Department of Medicine, Stony Brook University, Stony Brook, New York (G.M., C.C.W., Y.S., C.A.A., R.R., B.R.); and Medicon Pharmaceuticals, Inc. (P.P.C., B.R.), Stony Brook, New York
| | - Carol A Amella
- Department of Medicine, Stony Brook University, Stony Brook, New York (G.M., C.C.W., Y.S., C.A.A., R.R., B.R.); and Medicon Pharmaceuticals, Inc. (P.P.C., B.R.), Stony Brook, New York
| | - Robert Richards
- Department of Medicine, Stony Brook University, Stony Brook, New York (G.M., C.C.W., Y.S., C.A.A., R.R., B.R.); and Medicon Pharmaceuticals, Inc. (P.P.C., B.R.), Stony Brook, New York
| | - Panayiotis P Constantinides
- Department of Medicine, Stony Brook University, Stony Brook, New York (G.M., C.C.W., Y.S., C.A.A., R.R., B.R.); and Medicon Pharmaceuticals, Inc. (P.P.C., B.R.), Stony Brook, New York
| | - Basil Rigas
- Department of Medicine, Stony Brook University, Stony Brook, New York (G.M., C.C.W., Y.S., C.A.A., R.R., B.R.); and Medicon Pharmaceuticals, Inc. (P.P.C., B.R.), Stony Brook, New York
| |
Collapse
|
15
|
2-Dioleoyl-sn-glycero-3-phosphocholine-based nanoliposomes as an effective delivery platform for 17β-estradiol. Eur J Pharm Biopharm 2014; 86:369-75. [DOI: 10.1016/j.ejpb.2013.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 10/08/2013] [Accepted: 10/16/2013] [Indexed: 12/30/2022]
|
16
|
Mattheolabakis G, Rigas B, Constantinides PP. Nanodelivery strategies in cancer chemotherapy: biological rationale and pharmaceutical perspectives. Nanomedicine (Lond) 2013; 7:1577-90. [PMID: 23148540 DOI: 10.2217/nnm.12.128] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Nanotechnology is revolutionizing our approach to drug delivery, a key determinant of drug efficacy. Here, we present cancer drug delivery strategies that exploit nanotechnology, providing first an overview of tumor biology aspects that critically affect the design of drug delivery carriers, namely the enhanced permeability and retention effect, the lower tumor extracellular pH and tumor-specific antigens. In general, nanoscience-based approaches have circumvented limitations in the delivery of cancer therapeutics, related to their poor aqueous solubility and toxicity issues with conventional vehicles and resulted in improved pharmacokinetics and biodistribution. Included in the discussion are promising examples and pharmaceutical perspectives on liposomes, nanoemulsions, solid lipid nanoparticles, polymeric nanoparticles, dendrimers, carbon nanotubes and magnetic nanoparticles. As the cardinal features of the ideal multifunctional cancer drug nanocarrier are becoming clear, and drug development challenges are proactively addressed, we anticipate that future advances will enhance therapeutic outcomes by refining the delivery and targeting of complex payloads.
Collapse
Affiliation(s)
- George Mattheolabakis
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | | | | |
Collapse
|
17
|
Ouyang N, Ji P, Williams JL. A novel NSAID derivative, phospho-ibuprofen, prevents AOM-induced colon cancer in rats. Int J Oncol 2012; 42:643-50. [PMID: 23291777 PMCID: PMC3982714 DOI: 10.3892/ijo.2012.1756] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2012] [Accepted: 11/23/2012] [Indexed: 12/28/2022] Open
Abstract
The cancer chemopreventive properties and gastrointestinal toxicity of ibuprofen are well documented. Modification of existing NSAIDs has improved on the chemopreventive efficacy of this agent and reduced its toxicity. In this study, ibuprofen and a modified derivative (phospho-modified ibuprofen or p-ibuprofen) were used in a chemically induced model of colon cancer. Fisher 344 rats were injected with azoxymethane then treated with either ibuprofen (500 ppm) or p-ibuprofen (900 ppm) for 20 weeks to observe aberrant crypt foci (ACF) or 40 weeks to evaluate tumor incidence and multiplicity. β-catenin and p65 were measured in colonic tissues by immunofluorescence staining. Equal molar doses of ibuprofen (75 and 670 mg/kg) and p-ibuprofen (135 and 1,215 mg/kg) were administered to rats for 7 days to assess acute toxicity. The in vitro effect of p-ibuprofen on COX-2 and PGE(2) synthesis, β-catenin expression and NF-κB activity were examined in RAW 264.7 macrophage and HCT 116 colon cancer cells. At week 20, p-ibuprofen and ibuprofen significantly reduced the multiplicity of ACF compared with control (p<0.05); 31.2 and 37.9%, respectively. At week 40, p-ibuprofen and ibuprofen reduced the multiplicity of colon tumors compared with control (p<0.01) by 47.2 and 56.6%, respectively. Equal molar concentrations of ibuprofen (670 mg/kg) and p-ibuprofen (1,215 mg/kg) resulted in stomach ulceration in 85.7% (6 out of 7) and 14.3% (1 out of 7) of rats, respectively, with p<0.01. Immunofluoresence staining and western blot analysis demonstrated that both ibuprofen and p-ibuprofen suppressed β-catenin nuclear translocation in colon cancer cells. In addition, p-ibuprofen but not ibuprofen inhibited NF-κB activation in colon cancer cells. Collectively, these results suggest that p-ibuprofen is a potential effective novel drug for long-term use in colon cancer prevention.
Collapse
Affiliation(s)
- Nengtai Ouyang
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY 11794-8175, USA
| | | | | |
Collapse
|
18
|
Cheng KW, Mattheolabakis G, Wong CC, Ouyang N, Huang L, Constantinides PP, Rigas B. Topical phospho-sulindac (OXT-328) is effective in the treatment of non-melanoma skin cancer. Int J Oncol 2012; 41:1199-203. [PMID: 22842609 PMCID: PMC3583614 DOI: 10.3892/ijo.2012.1577] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 03/23/2012] [Indexed: 12/16/2022] Open
Abstract
Phospho-sulindac (P-S, OXT-328), a novel sulindac derivative, has shown superior anticancer efficacy and safety compared to sulindac. In this study, we investigated the efficacy of topical P-S hydrogel in the treatment of non-melanoma skin cancer in preclinical models. P-S is a potent inhibitor of A431 epidermoid carcinoma in vitro and achieves this effect by inhibiting cell proliferation and inducing apoptosis. The anticancer efficacy of topical and oral P-S was further evaluated in mice bearing A431 intradermal xenografts. Compared to the controls, topical P-S hydrogel inhibited the A431 xenografts by 70.5% (p<0.01), while oral P-S inhibited it by 43.4% (p<0.05), being significantly less effective than topical P-S (p= 0.017). Topical P-S hydrogel generated significant levels (>500 nmol/g tumor tissue) of intact P-S in the tumors, accounting for 92.5% of the total metabolites in the A431 xenografts. This local delivery of high levels of intact P-S to the A431 xenografts is an important contributor to the potent activity of topical P-S and no local or systemic side effects were noted in the treatment group. Thus, topical P-S is a promising treatment modality against non-melanoma skin cancer and merits further evaluation.
Collapse
Affiliation(s)
- Ka Wing Cheng
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | | | | | | | | | | | | |
Collapse
|