1
|
Basu A, Yang JY, Tsirukis VE, Loiacono A, Koch G, Khwaja IA, Krishnamurthy M, Fazio N, White E, Jha A, Shah S, Takmil C, Bagdas D, Demirer A, Master A, Natke E, Honkanen R, Huang L, Rigas B. Phosphosulindac (OXT-328) prevents and reverses chemotherapy induced peripheral neuropathy in mice. Front Neurosci 2024; 17:1240372. [PMID: 38347876 PMCID: PMC10860339 DOI: 10.3389/fnins.2023.1240372] [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: 06/14/2023] [Accepted: 10/19/2023] [Indexed: 02/15/2024] Open
Abstract
Background Chemotherapy-induced peripheral neuropathy (CIPN), a side effect of chemotherapy, is particularly difficult to treat. We explored whether phosphosulindac (PS), a modified NSAID, could treat CIPN. Methods CIPN was induced in male C57BL/6 J mice by paclitaxel, vincristine or oxaliplatin. Mechanical allodynia was measured with the von Frey test and cold allodynia with the acetone test. To determine the preventive effect of PS, it was administered 2 days before the induction of CIPN. Mouse Lewis lung carcinoma xenografts were used to determine if PS altered the chemotherapeutic efficacy of paclitaxel. Cultured cell lines were used to evaluate the effect of PS on neuroinflammation. Results Treatment with each of the three chemotherapeutic agents used to induce CIPN lowered the mechanical allodynia scores by 56 to 85% depending on the specific agent. PS gel was applied topically 3x/day for 16-22 days to the hind paws of mice with CIPN. This effect was dose-dependent. Unlike vehicle, PS returned mechanical allodynia scores back to pre-CIPN levels. PS had a similar effect on paclitaxel-induced CIPN cold allodynia. Sulindac, a metabolite of PS, had no effect on CIPN. PS significantly prevented CIPN compared to vehicle. Given concomitantly with paclitaxel to mice with lung cancer xenografts, PS relieved CIPN without affecting the anticancer effect of paclitaxel. The enantiomers of PS were equally efficacious against CIPN, suggesting the therapeutic suitability of the racemate PS. There were no apparent side effects of PS. PS suppressed the levels of IL-6, IL-10, CXCL1, and CXCL2 induced by paclitaxel in a neuroblastoma cell line, and macrophage activation to the M1 proinflammatory phenotype. Conclusion Topically applied PS demonstrated broad therapeutic and preventive efficacy against CIPN, preserved the anticancer effect of paclitaxel, and was safe. Its anti-CIPN effect appears to be mediated, in part, by suppression of neuroinflammation. These data support further evaluation of topical PS for the control of CIPN.
Collapse
Affiliation(s)
- Aryah Basu
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Jennifer Y. Yang
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Vasiliki E. Tsirukis
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Andrew Loiacono
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Gina Koch
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Ishan A. Khwaja
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Mahila Krishnamurthy
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Nicholas Fazio
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Emily White
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Aayushi Jha
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Shrila Shah
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Cameron Takmil
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Deniz Bagdas
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, United States
| | - Aylin Demirer
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, United States
| | - Adam Master
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Ernest Natke
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| | - Robert Honkanen
- Departments of Ophthalmology, Stony Brook University, Stony Brook, NY, United States
| | - Liqun Huang
- Medicon Pharmaceuticals, Inc, Setauket, NY, United States
| | - Basil Rigas
- Departments of Preventive Medicine, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
2
|
Huang W, Huang L, Wen Z, Honkanen RA, Rigas B. The Antiangiogenic Effect and Ocular Pharmacology of Novel Modified Nonsteroidal Anti-Inflammatory Drugs in the Treatment of Oxygen-Induced Retinopathy. J Ocul Pharmacol Ther 2023; 39:279-289. [PMID: 37172294 PMCID: PMC10178932 DOI: 10.1089/jop.2022.0113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2023] Open
Abstract
Purpose: To evaluate the hypothesis that 3 novel compounds, OXT-328, Q-922, and CL-717 show efficacy in the treatment of oxygen-induced retinopathy (OIR) and whether or not their route of administration is intravitreal, topical, or systemic. Methods: The OIR mouse model, characterized by an avascular area (AVA) and a neovascular area (NVA) of the retina, was used to study retinopathy of prematurity and other retinal diseases characterized by abnormal vessel growth. We measured the effect of our compounds on both the AVA and NVA in whole mounts of mouse retinal tissue. We also evaluated their ability to prevent new vessel formation in chicken chorioallantoic membranes (CAMs). Finally, we measured the in vitro uptake and biodistribution of topically applied CL-717 in human eye explants. Results: In mice with OIR, compared to controls, a single intravitreal administration of Q-922 or OXT-328 significantly reduced both AVA and NVA. CL-717 administered as eye drops over 5 days also reduced AVA and NVA, whereas OXT-328 eye drops had no effect. Q-922 given intraperitoneal (150 mg/kg/day × 5 days) reduced AVA and NVA. Remarkably, explanted human eyes bathed in CL-717 show rapid uptake and biodistribution in ocular tissues. In the chicken CAM model, all 3 compounds reduced the formation of new blood vessels by about one-third. No side effect in mice was observed, except for mild ocular surface irritation with Q-922. Conclusions: Systemic administration of Q-922 or topical administration of CL-717 holds particular promise for a simplified treatment of proliferative retinopathies without the necessity of intravitreal injections.
Collapse
Affiliation(s)
- Wei Huang
- Department of Ophthalmology, Population and Preventive Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Liqun Huang
- Medicon Pharmaceuticals, Inc., Setauket, New York, USA
| | - Ziyi Wen
- Medicon Pharmaceuticals, Inc., Setauket, New York, USA
| | - Robert A Honkanen
- Department of Ophthalmology, Population and Preventive Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Basil Rigas
- Department of Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
3
|
Hydrogel formulation of phosphosulindac allows once-a-day ocular dosing and limits its biodistribution to the anterior chamber: Application to dry eye disease treatment. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2021.102961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
4
|
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: 8.0] [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
|
5
|
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
|
6
|
The ocular pharmacokinetics and biodistribution of phospho-sulindac (OXT-328) formulated in nanoparticles: Enhanced and targeted tissue drug delivery. Int J Pharm 2019; 557:273-279. [DOI: 10.1016/j.ijpharm.2018.12.057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 12/14/2018] [Accepted: 12/21/2018] [Indexed: 12/18/2022]
|
7
|
A Second WNT for Old Drugs: Drug Repositioning against WNT-Dependent Cancers. Cancers (Basel) 2016; 8:cancers8070066. [PMID: 27429001 PMCID: PMC4963808 DOI: 10.3390/cancers8070066] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 06/24/2016] [Accepted: 07/07/2016] [Indexed: 12/21/2022] Open
Abstract
Aberrant WNT signaling underlies cancerous transformation and growth in many tissues, such as the colon, breast, liver, and others. Downregulation of the WNT pathway is a desired mode of development of targeted therapies against these cancers. Despite the urgent need, no WNT signaling-directed drugs currently exist, and only very few candidates have reached early phase clinical trials. Among different strategies to develop WNT-targeting anti-cancer therapies, repositioning of existing drugs previously approved for other diseases is a promising approach. Nonsteroidal anti-inflammatory drugs like aspirin, the anti-leprotic clofazimine, and the anti-trypanosomal suramin are among examples of drugs having recently revealed WNT-targeting activities. In total, 16 human-use drug compounds have been found to be working through the WNT pathway and show promise for their prospective repositioning against various cancers. Advances, hurdles, and prospects of developing these molecules as potential drugs against WNT-dependent cancers, as well as approaches for discovering new ones for repositioning, are the foci of the current review.
Collapse
|
8
|
Tsioulias GJ, Go MF, Rigas B. NSAIDs and Colorectal Cancer Control: Promise and Challenges. ACTA ACUST UNITED AC 2015; 1:295-301. [PMID: 26688785 DOI: 10.1007/s40495-015-0042-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The chemoprevention of colorectal cancer (CRC) is a realistic option given the low acceptance and cost of screening colonoscopy. NSAIDs, currently not recommended for CRC prevention, are the most promising agents. Here, we review relevant work and assess the chemopreventive potential of NSAIDs. The chemopreventive efficacy of NSAIDs is established by epidemiological and interventional studies as well as analyses of cardiovascular-prevention randomized clinical trials. The modest chemopreventive efficacy of NSAIDs is compounded by their significant toxicity that can be cumulative. Efforts to overcome these limitations include the use of drug combinations; the emphasis on the early stages of colon carcinogenesis such as aberrant crypt foci, which may require shorter periods of drug administration; and the development of several families of chemically modified NSAIDs such as derivatives of sulindac, nitro-NSAIDs and phospho-NSAIDs, with some of them appearing to have higher safety and efficacy than conventional NSAIDs and thus to be better candidate agents. The successful development of NSAIDs as chemopreventive agents will likely require a combination of the following: identification of subjects at high risk and/or those most likely to benefit from chemoprevention; optimization of the timing, dose and duration of administration of the chemopreventive agent; novel NSAID derivatives and/or combinations of agents; and agents that may prevent other diseases in addition to CRC. Ultimately, the clinical implementation of NSAIDs for the prevention of CRC will depend on a strategy that drastically shifts the currently unacceptable risk/benefit ratio in favor of chemoprevention.
Collapse
Affiliation(s)
- George J Tsioulias
- Department of Surgery, Medical Sciences Building G530, Rutgers Medical School of New Jersey, 185 South Orange Avenue, Newark, NJ 07103, Tel: 973-676-1000 x1801
| | - Mae F Go
- Gastroenterology Section, VA Southern Nevada Healthcare System, 6900 N. Pecos Rd, North Las Vegas, NV 89086, Tel: 702-791-9000
| | - Basil Rigas
- Stony Brook University, HSC, L4, Room 169, Stony Brook, NY 11794-8430, Tel: 631-638-2141
| |
Collapse
|
9
|
Rigas B, Tsioulias GJ. The evolving role of nonsteroidal anti-inflammatory drugs in colon cancer prevention: a cause for optimism. J Pharmacol Exp Ther 2015; 353:2-8. [PMID: 25589413 DOI: 10.1124/jpet.114.220806] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Colorectal cancer (CRC) is a serious yet preventable disease. The low acceptance and cost of colonoscopy as a screening method or CRC make chemoprevention an important option. Nonsteroidal anti-inflammatory drugs (NSAIDs), not currently recommended for CRC prevention, have the potential to evolve into the agents of choice for this indication. Here, we discuss the promise and challenge of NSAIDs for this chemopreventive application.Multiple epidemiologic studies, randomized clinical trials (RCTs) of sporadic colorectal polyp recurrence, RCTs in patients with hereditary colorectal cancer syndromes, and pooled analyses of cardiovascular-prevention RCTs linked to cancer outcomes have firmly established the ability of conventional NSAIDs to prevent CRC. NSAIDs, however, are seriously limited by their toxicity,which can become cumulative with their long-term administration for chemoprevention, whereas drug interactions in vulnerable elderly patients compound their safety. Newer, chemically modified NSAIDs offer the hope of enhanced efficacy and safety.Recent work also indicates that targeting earlier stages of colorectal carcinogenesis, such as the lower complexity aberrant crypt foci, is a promising approach that may only require relatively short use of chemopreventive agents. Drug combination approaches exemplified by sulindac plus difluoromethylornithine appear very efficacious. Identification of those at risk or most likely to benefit from a given intervention using predictive biomarkers may usher in personalized chemoprevention. Agents that offer simultaneous chemoprevention of diseases in addition to CRC, e.g., cardiovascular and/or neurodegenerative diseases,may have a much greater potential for a broad clinical application.
Collapse
Affiliation(s)
- Basil Rigas
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA. basil.rigas@stonybrookmedicine
| | | |
Collapse
|
10
|
Wong CC, Cheng KW, Papayannis I, Mattheolabakis G, Huang L, Xie G, Ouyang N, Rigas B. Phospho-NSAIDs have enhanced efficacy in mice lacking plasma carboxylesterase: implications for their clinical pharmacology. Pharm Res 2014; 32:1663-75. [PMID: 25392229 DOI: 10.1007/s11095-014-1565-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 10/28/2014] [Indexed: 12/31/2022]
Abstract
PURPOSE The purpose of the study was to evaluate the metabolism, pharmacokinetics and efficacy of phospho-NSAIDs in Ces1c-knockout mice. METHODS Hydrolysis of phospho-NSAIDs by Ces1c was investigated using Ces1c-overexpressing cells. The rate of phospho-NSAID hydrolysis was compared between wild-type, Ces1c+/- and Ces1c-/- mouse plasma in vitro, and the effect of plasma Ces1c on the cytotoxicity of phospho-NSAIDs was evaluated. Pharmacokinetics of phospho-sulindac was examined in wild-type and Ces1c-/- mice. The impact of Ces1c on the efficacy of phospho-sulindac was investigated using lung and pancreatic cancer models in vivo. RESULTS Phospho-NSAIDs were extensively hydrolyzed in Ces1c-overexpressing cells. Phospho-NSAID hydrolysis in wild-type mouse plasma was 6-530-fold higher than that in the plasma of Ces1c-/- mice. Ces1c-expressing wild-type mouse serum attenuated the in vitro cytotoxicity of phospho-NSAIDs towards cancer cells. Pharmacokinetic studies of phospho-sulindac using wild-type and Ces1c-/- mice demonstrated 2-fold less inactivation of phospho-sulindac in the latter. Phospho-sulindac was 2-fold more efficacious in inhibiting the growth of lung and pancreatic carcinoma in Ces1c -/- mice, as compared to wild-type mice. CONCLUSIONS Our results indicate that intact phospho-NSAIDs are the pharmacologically active entities and phospho-NSAIDs are expected to be more efficacious in humans than in rodents due to their differential expression of carboxylesterases.
Collapse
Affiliation(s)
- Chi C Wong
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, HSC, T-17 Room 080, Stony Brook, NY, 11794-8173, USA
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Suthar SK, Sharma M. Recent Developments in Chimeric NSAIDs as Safer Anti-Inflammatory Agents. Med Res Rev 2014; 35:341-407. [DOI: 10.1002/med.21331] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Sharad Kumar Suthar
- Department of Pharmacy; Jaypee University of Information Technology; Waknaghat 173234 India
| | - Manu Sharma
- Department of Pharmacy; Jaypee University of Information Technology; Waknaghat 173234 India
| |
Collapse
|
12
|
Xie G, Cheng KW, Huang L, Rigas B. The in vitro metabolism of phospho-sulindac amide, a novel potential anticancer agent. Biochem Pharmacol 2014; 91:249-55. [PMID: 25044307 DOI: 10.1016/j.bcp.2014.07.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 07/09/2014] [Accepted: 07/11/2014] [Indexed: 12/21/2022]
Abstract
Phospho-sulindac amide (PSA) is a novel potential anti-cancer and anti-inflammatory agent. Here we report the metabolism of PSA in vitro. PSA was rapidly hydroxylated at its butane-phosphate moiety to form two di-hydroxyl-PSA and four mono-hydroxyl-PSA metabolites in mouse and human liver microsomes. PSA also can be oxidized or reduced at its sulindac moiety to form PSA sulfone and PSA sulfide, respectively. PSA was mono-hydroxylated and cleared more rapidly in mouse liver microsomes than in human liver microsomes. Of eight major human cytochrome P450s (CYPs), CYP3A4 and CYP2D6 exclusively catalyzed the hydroxylation and sulfoxidation reactions of PSA, respectively. We also examined the metabolism of PSA by three major human flavin monooxygenases (FMOs). FMO1, FMO3 and FMO5 were all capable of catalyzing the sulfoxidation (but not hydroxylation) of PSA, with FMO1 being by far the most active isoform. PSA was predominantly sulfoxidized in human kidney microsomes because FMO1 is the dominant isoform in human kidney. PSA (versus sulindac) is a preferred substrate of both CYPs and FMOs, likely because of its greater lipophilicity and masked-COOH group. Ketoconazole (a CYP3A4 inhibitor) and alkaline pH strongly inhibited the hydroxylation of PSA, but moderately suppressed its sulfoxidation in liver microsomes. Together, our results establish the metabolic pathways of PSA, identify the major enzymes mediating its biotransformations and reveal significant inter-species and inter-tissue differences in its metabolism.
Collapse
Affiliation(s)
- Gang Xie
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Ka-Wing Cheng
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Liqun Huang
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Basil Rigas
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; Medicon Pharmaceuticals, Inc., Stony Brook, NY 11790, USA.
| |
Collapse
|
13
|
MURRAY ONIKAT, WONG CHIC, VRANKOVA KVETOSLAVA, RIGAS BASIL. Phospho-sulindac inhibits pancreatic cancer growth: NFATc1 as a drug resistance candidate. Int J Oncol 2014; 44:521-9. [PMID: 24284479 PMCID: PMC3898803 DOI: 10.3892/ijo.2013.2190] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 10/07/2013] [Indexed: 12/13/2022] Open
Abstract
Phospho-sulindac (P-S), a promising anticancer agent, is efficacious in pre-clinical models of human cancer and is apparently safe. Here, we studied the effect of P-S on pancreatic cancer growth. We found that P-S strongly inhibits the growth of human pancreatic cancer cells in vitro, is efficacious in inhibiting the growth of pancreatic xenografts in nude mice, and has an excellent safety profile. Microarray analysis revealed that P-S induced the expression of nuclear factor of activated T-cells, isoform c1 (NFATc1) gene. NFATc1, a calcineurin-responsive transcription factor associated with aggressive pancreatic cancer. The role of increased NFATc1 expression on the growth inhibitory effect of P-S on cancer growth was evaluated by silencing or by overexpressing it both in vitro and in vivo. We found that when the expression of NFATc1 was abrogated by RNAi, pancreatic cancer cells were more responsive to treatment with P-S. Conversely, overexpressing the NFATc1 gene made the pancreatic cancer cells less responsive to treatment with P-S. NFATc1 likely mediates drug resistance to P-S and is an unfavorable prognostic factor that predicts poor tumor response. We also demonstrated that NFATc1-mediated resistance can be overcome by cyclosporin A (CsA), an NFAT inhibitor, and that the combination of P-S and CsA synergistically inhibited pancreatic cancer cell growth. In conclusion, our preclinical data establish P-S as an efficacious drug for pancreatic cancer in preclinical models, which merits further evaluation.
Collapse
Affiliation(s)
- ONIKA T. MURRAY
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY 11794-8173,
USA
| | - CHI C. WONG
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY 11794-8173,
USA
| | - KVETOSLAVA VRANKOVA
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY 11794-8173,
USA
| | - BASIL RIGAS
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY 11794-8173,
USA
| |
Collapse
|
14
|
Cheng KW, Wong CC, Mattheolabakis G, Xie G, Huang L, Rigas B. Curcumin enhances the lung cancer chemopreventive efficacy of phospho-sulindac by improving its pharmacokinetics. Int J Oncol 2013; 43:895-902. [PMID: 23807084 PMCID: PMC3787887 DOI: 10.3892/ijo.2013.1995] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2013] [Accepted: 04/29/2013] [Indexed: 12/17/2022] Open
Abstract
Phospho-sulindac (PS) is a safe sulindac derivative with promising anticancer efficacy in colon cancer. We evaluated whether its combination with curcumin could enhance the efficacy in the treatment of lung cancer. Curcumin, the principal bioactive component in turmeric, has demonstrated versatile capabilities to modify the therapeutic efficacy of a wide range of anticancer agents. Here, we evaluated the effect of co-administration of curcumin on the anticancer activity of PS in a mouse xenograft model of human lung cancer. Curcumin enhanced the cellular uptake of PS in human lung and colon cancer cell lines. To assess the potential synergism between curcumin and PS in vivo, curcumin was suspended in 10% Tween-80 or formulated in micellar nanoparticles and given to mice by oral gavage prior to the administration of PS. Both formulations of curcumin significantly improved the pharmacokinetic profiles of PS, with the 10% Tween-80 suspension being much more effective than the nanoparticle formation. However, curcumin did not exhibit any significant modification of the metabolite profile of PS. Furthermore, in a mouse subcutaneous xenograft model of human lung cancer, PS (200 mg/kg) in combination with curcumin (500 mg/kg) suspended in 10% Tween-80 (51% inhibition, p<0.05) was significantly more efficacious than PS plus micelle curcumin (30%) or PS (25%) or curcumin alone (no effect). Consistent with the improved pharmacokinetics, the combination treatment group had higher levels of PS and its metabolites in the xenografts compared to PS alone. Our results show that curcumin substantially improves the pharmacokinetics of PS leading to synergistic inhibition of the growth of human lung cancer xenografts, representing a promising drug combination.
Collapse
Affiliation(s)
- Ka-Wing Cheng
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, NY 11794-8173, USA
| | | | | | | | | | | |
Collapse
|
15
|
Cheng KW, Wong CC, Alston N, Mackenzie GG, Huang L, Ouyang N, Xie G, Wiedmann T, Rigas B. Aerosol administration of phospho-sulindac inhibits lung tumorigenesis. Mol Cancer Ther 2013; 12:1417-28. [PMID: 23645590 DOI: 10.1158/1535-7163.mct-13-0006-t] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Phospho-sulindac is a sulindac derivative with promising anticancer activity in lung cancer, but its limited metabolic stability presents a major challenge for systemic therapy. We reasoned that inhalation delivery of phospho-sulindac might overcome first-pass metabolism and produce high levels of intact drug in lung tumors. Here, we developed a system for aerosolization of phospho-sulindac and evaluated the antitumor efficacy of inhaled phospho-sulindac in an orthotopic model of human non-small cell lung cancer (A549 cells). We found that administration by inhalation delivered high levels of phospho-sulindac to the lungs and minimized its hydrolysis to less active metabolites. Consequently, inhaled phospho-sulindac (6.5 mg/kg) was highly effective in inhibiting lung tumorigenesis (75%; P < 0.01) and significantly improved the survival of mice bearing orthotopic A549 xenografts. Mechanistically, phospho-sulindac suppressed lung tumorigenesis by (i) inhibiting EGF receptor (EGFR) activation, leading to profound inhibition of Raf/MEK/ERK and PI3K/AKT/mTOR survival cascades; (ii) inducing oxidative stress, which provokes the collapse of mitochondrial membrane potential and mitochondria-dependent cell death; and (iii) inducing autophagic cell death. Our data establish that inhalation delivery of phospho-sulindac is an efficacious approach to the control of lung cancer, which merits further evaluation.
Collapse
Affiliation(s)
- Ka Wing Cheng
- Division of Cancer Prevention, Stony Brook University, Stony Brook, NY 11794, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Mattheolabakis G, Mackenzie GG, Huang L, Ouyang N, Cheng KW, Rigas B. Topically applied phospho-sulindac hydrogel is efficacious and safe in the treatment of experimental arthritis in rats. Pharm Res 2013; 30:1471-82. [PMID: 23483440 DOI: 10.1007/s11095-012-0953-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 12/03/2012] [Indexed: 12/26/2022]
Abstract
PURPOSE Formulate phospho-sulindac (P-S, OXT-328) in a Pluronic hydrogel to be used as a topical anti-inflammatory agent and study its efficacy, safety and pharmacokinetics in an arthritis model. METHODS LEW/crlBR rats with Freund's adjuvant-induced arthritis were treated with P-S formulated in Pluronic hydrogel (PSH). We determined the clinical manifestations of arthritis including the locomotor activity of the rats; evaluated joints for inflammation, bone resorption, cartilage damage, COX-2 expression and NF-κB activation; assayed plasma IL-6 and IL-10 levels; and studied the pharmacokinetics of P-S in rats after topical or oral administration. RESULTS PSH applied at the onset of arthritis or when arthritis was fully developed, suppressed it by 56-82%, improved the locomotor activity of the rats 2.1-4.4 fold, suppressed synovial inflammation, bone resorption, cartilage damage, NF-κB activation and COX-2 expression but not plasma IL-6 and IL-10 levels. There were no side effects. PSH produced rapidly high local levels of P-S with <14% of P-S reaching the circulation, while orally administered P-S was rapidly metabolized generating much lower joint levels of P-S. CONCLUSIONS Topical application of PSH is efficacious and safe in the treatment of Freund's adjuvant-induced arthritis; has a favorable pharmacokinetic profile; and likely acts by suppressing key pro-inflammatory signaling pathways.
Collapse
Affiliation(s)
- George Mattheolabakis
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, HSC T17-080, Stony Brook, New York 11794-8173, USA
| | | | | | | | | | | |
Collapse
|
17
|
Xie G, Wong CC, Cheng KW, Huang L, Constantinides PP, Rigas B. Regioselective oxidation of phospho-NSAIDs by human cytochrome P450 and flavin monooxygenase isoforms: implications for their pharmacokinetic properties and safety. Br J Pharmacol 2013; 167:222-32. [PMID: 22489789 DOI: 10.1111/j.1476-5381.2012.01982.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Phospho-ibuprofen (MDC-917) and phospho-sulindac (OXT-328) are highly effective in cancer and arthritis treatment in preclinical models. Here, we investigated their metabolism by major human cytochrome P450s (CYPs) and flavin monooxygenases (FMOs). EXPERIMENTAL APPROACH The CYP/FMO-catalysed metabolism of phospho-ibuprofen and phospho-sulindac was studied by using in silico prediction modelling and a direct experimental approach. KEY RESULTS The CYP isoforms catalyse the oxidation of non-steroidal anti-inflammatory drugs (NSAIDs) and phospho-NSAIDs, with distinct activity and regioselectivity. CYP1A2, 2C19, 2D6 and 3A4 oxidize phospho-ibuprofen, but not ibuprofen; whereas CYP2C9 oxidizes ibuprofen, but not phospho-ibuprofen. All CYPs tested oxidize phospho-sulindac, but not sulindac. Among the five CYPs evaluated, CYP3A4 and 2D6 are the most active in the oxidation of phospho-ibuprofen and phospho-sulindac respectively. FMOs oxidized phospho-sulindac and sulindac, but not phospho-ibuprofen or ibuprofen. FMOs were more active towards phospho-sulindac than sulindac, indicating that phospho-sulindac is a preferred substrate of FMOs. The susceptibility of phospho-NSAIDs to CYP/FMO-mediated metabolism was also reflected in their rapid oxidation by human and mouse liver microsomes, which contain a full complement of CYPs and FMOs. Compared with conventional NSAIDs, the higher activity of CYPs towards phospho-ibuprofen and phospho-sulindac may be due to their greater lipophilicity, a key parameter for CYP binding. CONCLUSIONS AND IMPLICATIONS CYPs and FMOs play an important role in the metabolism of phospho-NSAIDs, resulting in differential pharmacokinetic profiles between phospho-NSAIDs and NSAIDs in vivo. The consequently more rapid detoxification of phospho-NSAIDs is likely to contribute to their greater safety.
Collapse
Affiliation(s)
- Gang Xie
- Department of Medicine, Division of Cancer Prevention, Stony Brook University, Stony Brook, NY 11794-8173, USA
| | | | | | | | | | | |
Collapse
|
18
|
Cheng TJR, Weinheimer S, Tarbet EB, Jan JT, Cheng YSE, Shie JJ, Chen CL, Chen CA, Hsieh WC, Huang PW, Lin WH, Wang SY, Fang JM, Hu OYP, Wong CH. Development of oseltamivir phosphonate congeners as anti-influenza agents. J Med Chem 2012; 55:8657-70. [PMID: 23009169 PMCID: PMC3492761 DOI: 10.1021/jm3008486] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Oseltamivir phosphonic acid (tamiphosphor, 3a), its monoethyl ester (3c), guanidino-tamiphosphor (4a), and its monoethyl ester (4c) are potent inhibitors of influenza neuraminidases. They inhibit the replication of influenza viruses, including the oseltamivir-resistant H275Y strain, at low nanomolar to picomolar levels, and significantly protect mice from infection with lethal doses of influenza viruses when orally administered with 1 mg/kg or higher doses. These compounds are stable in simulated gastric fluid, liver microsomes, and human blood and are largely free from binding to plasma proteins. Pharmacokinetic properties of these inhibitors are thoroughly studied in dogs, rats, and mice. The absolute oral bioavailability of these compounds was lower than 12%. No conversion of monoester 4c to phosphonic acid 4a was observed in rats after intravenous administration, but partial conversion of 4c was observed with oral administration. Advanced formulation may be investigated to develop these new anti-influenza agents for better therapeutic use.
Collapse
MESH Headings
- Acetamides/chemical synthesis
- Acetamides/pharmacokinetics
- Acetamides/pharmacology
- Administration, Oral
- Animals
- Antiviral Agents/chemical synthesis
- Antiviral Agents/pharmacokinetics
- Antiviral Agents/pharmacology
- Biological Availability
- Blood Proteins/metabolism
- Cyclohexenes/chemical synthesis
- Cyclohexenes/pharmacokinetics
- Cyclohexenes/pharmacology
- Cytopathogenic Effect, Viral/drug effects
- Dogs
- Drug Resistance, Viral
- Drug Stability
- Female
- Humans
- Influenza A Virus, H1N1 Subtype/drug effects
- Influenza A Virus, H1N1 Subtype/enzymology
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H5N1 Subtype/drug effects
- Influenza A Virus, H5N1 Subtype/enzymology
- Alphainfluenzavirus/drug effects
- Alphainfluenzavirus/enzymology
- Alphainfluenzavirus/genetics
- Betainfluenzavirus/drug effects
- Betainfluenzavirus/enzymology
- Madin Darby Canine Kidney Cells
- Male
- Mice
- Mice, Inbred BALB C
- Microsomes, Liver/metabolism
- Mutation
- Neuraminidase/antagonists & inhibitors
- Orthomyxoviridae Infections/drug therapy
- Oseltamivir/pharmacology
- Phosphorous Acids
- Protein Binding
- Rats
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Ting-Jen R. Cheng
- The Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Road, Taipei 11529, Taiwan
| | - Steven Weinheimer
- TaiMed Biologics, 5251 California Avenue, Suite 230, Irvine, CA 92617, United States
| | - E. Bart Tarbet
- Institute for Antiviral Research, Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, Utah 84322, United States
| | - Jia-Tsrong Jan
- The Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Road, Taipei 11529, Taiwan
| | - Yih-Shyun E. Cheng
- The Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Road, Taipei 11529, Taiwan
| | - Jiun-Jie Shie
- The Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Road, Taipei 11529, Taiwan
| | - Chun-Lin Chen
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei 106, Taiwan
| | - Chih-An Chen
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei 106, Taiwan
| | - Wei-Che Hsieh
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei 106, Taiwan
| | - Pei-Wei Huang
- School of Pharmacy, National Defense Medical Center, No. 161, Sec. 6, Minquan E. Rd., Taipei 114, Taiwan
| | - Wen-Hao Lin
- School of Pharmacy, National Defense Medical Center, No. 161, Sec. 6, Minquan E. Rd., Taipei 114, Taiwan
| | - Shi-Yun Wang
- The Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Road, Taipei 11529, Taiwan
| | - Jim-Min Fang
- The Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Road, Taipei 11529, Taiwan
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei 106, Taiwan
| | - Oliver Yoa-Pu Hu
- School of Pharmacy, National Defense Medical Center, No. 161, Sec. 6, Minquan E. Rd., Taipei 114, Taiwan
| | - Chi-Huey Wong
- The Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Road, Taipei 11529, Taiwan
| |
Collapse
|
19
|
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: 1.2] [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
|
20
|
In vitro and in vivo metabolic studies of phospho-aspirin (MDC-22). Pharm Res 2012; 29:3292-301. [PMID: 22782648 DOI: 10.1007/s11095-012-0821-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 06/22/2012] [Indexed: 12/21/2022]
Abstract
PURPOSE To investigate the metabolism of phospho-aspirin (PA, MDC-22), a novel anti-cancer and anti-inflammatory agent. METHODS The metabolism of PA was studied in the liver and intestinal microsomes from mouse, rat and human. RESULTS PA is rapidly deacetylated to phospho-salicylic acid (PSA), which undergoes regioselective oxidation to generate 3-OH-PSA and 5-OH-PSA. PSA also can be hydrolyzed to give salicylic acid (SA), which can be further glucuronidated. PA is far more stable in human liver or intestinal microsomes compared to those from mouse or rat due to its slowest deacetylation in human microsomes. Of the five major human cytochrome P450 (CYP) isoforms, CYP2C19 and 2D6 are the most active towards PSA. In contrast to PSA, conventional SA is not appreciably oxidized by the CYPs and liver microsomes, indicating that PSA is a preferred substrate of CYPs. Similarly, PA, in contrast to PSA, cannot be directly oxidized by CYPs and liver microsomes, indicating that the acetyl group of PA abrogates its oxidation by CYPs. CONCLUSIONS Our findings establish the metabolism of PA, reveal significant inter-species differences in its metabolic transformations, and provide an insight into the role of CYPs in these processes.
Collapse
|
21
|
Phospho-sulindac (OXT-328) inhibits the growth of human lung cancer xenografts in mice: enhanced efficacy and mitochondria targeting by its formulation in solid lipid nanoparticles. Pharm Res 2012; 29:3090-101. [PMID: 22723123 DOI: 10.1007/s11095-012-0801-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 06/06/2012] [Indexed: 12/27/2022]
Abstract
PURPOSE To evaluate the antitumor efficacy of solid lipid nanoparticle-encapsulated phospho-sulindac (SLN-PS) in human lung cancer. METHODS PS was incorporated into SLNs using the emulsion evaporation technique. We determined the antitumor activity of SLN-PS in cultured lung cancer cells. The performance of SLN-PS was further evaluated by pharmacokinetic studies in mice and in a model of human lung cancer xenografts in nude mice. RESULTS SLN-PS was >4-fold more potent than PS in inhibiting the growth of A549 and H510 cells in vitro. SLN-PS enhanced cellular uptake and facilitated PS accumulation in mitochondria, leading to oxidative stress and apoptosis via the mitochondrial-apoptosis pathway. SLN-PS was highly effective in suppressing the growth of A549 xenografts (78% inhibition compared to control, p < 0.01); while PS had no significant effect. Formulation of PS in SLNs resulted in improved pharmacokinetics in mice and an enhanced (≈ 14-fold) accumulation of PS and its metabolites in A549 xenografts. Finally, SLN-PS enhanced urinary F2-isoprostane uniquely in mice bearing A549 xenografts compared to untreated controls, suggesting that SLN-PS specifically induced oxidative stress in tumors. CONCLUSIONS Our results show that SLN-PS is efficacious in suppressing the growth of lung cancer and merits further evaluation.
Collapse
|
22
|
Wong CC, Cheng KW, Xie G, Zhou D, Zhu CH, Constantinides PP, Rigas B. Carboxylesterases 1 and 2 hydrolyze phospho-nonsteroidal anti-inflammatory drugs: relevance to their pharmacological activity. J Pharmacol Exp Ther 2011; 340:422-32. [PMID: 22085648 DOI: 10.1124/jpet.111.188508] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Phospho-nonsteroidal anti-inflammatory drugs (phospho-NSAIDs) are novel NSAID derivatives with improved anticancer activity and reduced side effects in preclinical models. Here, we studied the metabolism of phospho-NSAIDs by carboxylesterases and assessed the impact of carboxylesterases on the anticancer activity of phospho-NSAIDs in vitro and in vivo. The expression of human liver carboxylesterase (CES1) and intestinal carboxylesterase (CES2) in human embryonic kidney 293 cells resulted in the rapid intracellular hydrolysis of phospho-NSAIDs. Kinetic analysis revealed that CES1 is more active in the hydrolysis of phospho-sulindac, phospho-ibuprofen, phospho-naproxen, phospho-indomethacin, and phospho-tyrosol-indomethacin that possessed a bulky acyl moiety, whereas the phospho-aspirins are preferentially hydrolyzed by CES2. Carboxylesterase expression leads to a significant attenuation of the in vitro cytotoxicity of phospho-NSAIDs, suggesting that the integrity of the drug is critical for anticancer activity. Benzil and bis-p-nitrophenyl phosphate (BNPP), two carboxylesterase inhibitors, abrogated the effect of carboxylesterases and resensitized carboxylesterase-expressing cells to the potent cytotoxic effects of phospho-NSAIDs. In mice, coadministration of phospho-sulindac and BNPP partially protected the former from esterase-mediated hydrolysis, and this combination more effectively inhibited the growth of AGS human gastric xenografts in nude mice (57%) compared with phospho-sulindac alone (28%) (p = 0.037). Our results show that carboxylesterase mediates that metabolic inactivation of phospho-NSAIDs, and the inhibition of carboxylesterases improves the efficacy of phospho-NSAIDs in vitro and in vivo.
Collapse
Affiliation(s)
- Chi C Wong
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, New York 11794-8173, USA
| | | | | | | | | | | | | |
Collapse
|