Gastroenterology

Gastroenterology

Volume 139, Issue 4, October 2010, Pages 1320-1332
Gastroenterology

Basic—Alimentary Tract
Phospho-Sulindac (OXT-328), a Novel Sulindac Derivative, Is Safe and Effective in Colon Cancer Prevention in Mice

https://doi.org/10.1053/j.gastro.2010.06.044Get rights and content

Background & Aims

Nonsteroidal anti-inflammatory drugs (NSAIDs) are effective cancer chemopreventive agents. However, chronic administration of NSAIDs is associated with significant side effects, mainly of the gastrointestinal tract. Given these limitations, we synthesized phospho-sulindac (P-S; OXT-328), a novel sulindac derivative.

Methods

Here, we evaluated the safety and efficacy of P-S in preclinical models, including its mechanism of action with human colon cancer cell (HCCC) lines and animal tumor models.

Results

(1) Compared with sulindac, P-S is much more potent in inhibiting the growth of cultured HCCCs and more efficacious in preventing the growth of HT-29 xenografts in nude mice. P-S also prevents the growth of intestinal tumors in Apc/Min mice. (2) In combination with difluoromethylornithine (DFMO), P-S reduced tumor multiplicity in Apc/Min mice by 90%. (3) P-S is much safer than sulindac as evidenced by its in vitro toxicologic evaluation and animal toxicity studies. Mechanistically, P-S increases the intracellular levels of reactive oxygen and nitrogen species, which are key early mediators of its chemopreventive effect. Moreover, P-S induces spermidine/spermine N1-acetyltransferase enzymatic activity, and together with DFMO it reduces polyamine levels in vitro and in vivo.

Conclusions

P-S displays considerable safety and efficacy, two pharmacologic properties that are essential for a potential cancer chemopreventive agent, and thus merits further evaluation.

Section snippets

Reagents and Cell Culture

P-S was synthesized as reported.6 Sulindac, A23187, and N-acetyl-L-cysteine (NAC) were from Sigma. Human colon cancer cell lines and the normal human colon cell line NCM460 were grown as monolayer as suggested by American Type Culture Collection (Manassas, VA). Cell growth was determined with the MTT assay (Promega, Madison, WI) following the manufacturer's instructions.7 Cells were treated with P-S with or without DFMO for ≤48 hours. Cell proliferation was assayed by 5-bromo-2′-deoxyuridine

P-S Inhibits the Growth of Cultured Colon Cancer Cells Through a Strong Cytokinetic Effect

To study the effect of P-S and sulindac on cell growth, we determined in various human colon cancer cells the 24-hour concentration values that inhibit their growth by 50% (IC50; Figure 1B). The IC50 values of P-S varied little among these cell lines (70–79.3 μmol/L), whereas those of sulindac were consistently >1000 μmol/L, indicating a potency enhancement of >14.2-fold.

We then elucidated the underlying cytokinetic effect of P-S on HT-29 and SW480 cells. P-S markedly reduced

Discussion

The successful pharmacologic agent against cancer must be effective and must lack significant side effects. Here, we demonstrate that P-S seems to meet both these requirements, markedly inhibiting intestinal tumors by itself and to a greater extent in combination with DFMO and appearing much safer than sulindac in preclinical studies.

P-S is a potent inhibitor of colon cancer cell lines (>14-fold more potent than sulindac) and exerts a profound inhibitory effect in preclinical models of colon

References (17)

There are more references available in the full text version of this article.

Cited by (51)

  • Phospho-Aspirin (MDC-22) inhibits pancreatic cancer growth in patient-derived tumor xenografts and KPC mice by targeting EGFR: Enhanced efficacy in combination with irinotecan

    2022, Neoplasia (United States)
    Citation Excerpt :

    We have previously identified MDC-22 (Fig. 1A) as a potent EGFR inhibitor [13,14], and documented that MDC-22 possesses strong chemopreventive properties in PC [14]. This novel agent has been synthesized based on a general approach where a specific chemical modification of known drugs enhances their desired anticancer properties, mainly their safety and efficacy [15,16]. In the present study, we examined the therapeutic efficacy of MDC-22 in two clinically relevant models of PC.

  • The ocular pharmacokinetics and biodistribution of phospho-sulindac (OXT-328) formulated in nanoparticles: Enhanced and targeted tissue drug delivery

    2019, International Journal of Pharmaceutics
    Citation Excerpt :

    Phospho-sulindac (PS, OXT-328) is a novel agent with anticancer and anti-inflammatory properties. The anticancer properties of PS encompass a variety of tumors, including colon, lung, non-melanoma skin cancer and pancreatic cancer (Cheng et al., 2018; Cheng et al., 2012, 2013; Mackenzie et al., 2010; Murray et al., 2014; Zhu et al., 2012). The anti-inflammatory efficacy of PS has been demonstrated in an animal model of arthritis, in which it was applied topically formulated in a hydrogel and reversed large joint arthritis (Mattheolabakis et al., 2013).

  • Phosphosulindac is efficacious in an improved concanavalin A-based rabbit model of chronic dry eye disease

    2018, Translational Research
    Citation Excerpt :

    Because the ideal treatment for DED is not currently available, the development of new treatments for DED is an active area of research. Phosphosulindac (PS, OXT-328) is a small molecule whose salient features, based on extensive preclinical work, are its pharmacologic efficacy and outstanding safety.26-28 PS is not a prodrug of the NSAID sulindac as the entire PS molecule is required for its pharmacologic activity.29,30

  • A novel ibuprofen derivative with anti-lung cancer properties: Synthesis, formulation, pharmacokinetic and efficacy studies

    2014, International Journal of Pharmaceutics
    Citation Excerpt :

    This effect was noted both in cultured cells, by using the ROS probe DCF-DA, and in animals, by assaying urinary levels of 8-iso-PGF2α, a widely used marker of oxidative stress in vivo (Milne et al., 2005). This finding is in agreement with previous studies (Huang et al., 2010; Mackenzie et al., 2010, 2011), which showed that ROS induction is a pivotal event in the anticancer effect of phospho-NSAIDs. Cancer cells are often in a heightened state of oxidative stress (Diehn et al., 2009).

  • NSAID-induced corneal melt: Clinical importance, pathogenesis, and risk mitigation

    2020, Survey of Ophthalmology
    Citation Excerpt :

    Cell death seems to be a primary event, and it is likely that “compromised” corneal epithelial cells respond differently to NSAIDs than healthy ones. Indeed, differential response to NSAIDs between normal and abnormal cells in terms of cell death is known.55 The persistence of the damage to the corneal surface (diminished tissue repair) appears to be a fateful development that, with the help of the right MMPs breaking down collagen, can culminate in the catastrophic perforation of the globe.

View all citing articles on Scopus

Conflicts of interest The authors disclose no conflicts, except for B.R., who has an equity position in Medicon, Inc.

Funding This study was supported by grant R01-CA139453; N01-CN-43302 WA#7.

View full text