Small Molecule
C20H17FO4S
59973-80-7
Exisulind, known chemically as sulindac sulfone, is the primary oxidative metabolite of the non-steroidal anti-inflammatory drug (NSAID) sulindac.[1] It was developed as the lead compound in a novel class of agents termed Selective Apoptotic Anti-neoplastic Drugs (SAANDs), distinguished by an anticancer mechanism deliberately separated from the cyclooxygenase (COX) inhibition characteristic of its parent compound.[2] The core mechanism of Exisulind involves the inhibition of cyclic guanosine monophosphate phosphodiesterases (cGMP-PDEs), particularly isoforms PDE2 and PDE5.[5] This inhibition leads to a sustained elevation of intracellular cGMP, which in turn activates protein kinase G (PKG). Activated PKG promotes programmed cell death (apoptosis) through downstream pathways, including the degradation of oncogenic β-catenin and activation of the c-Jun NH2-terminal kinase (JNK) pathway.[5]
The drug underwent an extensive clinical development program investigating its potential as both a chemopreventive and therapeutic agent across several cancers. The most promising signals of efficacy were observed in familial adenomatous polyposis (FAP) and prostate cancer. In FAP, clinical trials demonstrated that Exisulind could significantly reduce the formation of precancerous colorectal polyps.[3] In patients with biochemically recurrent prostate cancer, it significantly slowed the rate of rise in prostate-specific antigen (PSA) levels.[9] In contrast, multiple combination trials in non-small cell lung cancer (NSCLC) failed to show any benefit over standard chemotherapy, despite strong preclinical evidence of synergy.[10]
Ultimately, the clinical development of Exisulind for its primary indications was halted. Despite promising efficacy, the drug exhibited a narrow therapeutic window, with dose-limiting hepatotoxicity emerging as the critical safety concern.[12] The dose required for consistent therapeutic effect was too close to the dose that caused unacceptable, albeit reversible, liver enzyme elevations, rendering its risk-benefit profile unfavorable for long-term chemoprevention. This led to the U.S. Food and Drug Administration (FDA) issuing a "non-approvable" letter for the FAP indication in 2000.[3]
Despite these setbacks, Exisulind remains a molecule of significant scientific interest. Its development journey offers critical lessons in targeted therapy and the challenges of clinical translation. More recently, its unique mechanism of action has led to a potential new application. In 2014, the European Medicines Agency (EMA) granted Exisulind orphan designation for the treatment of familial cerebral cavernous malformations (FCCM), a rare vascular disease also linked to β-catenin dysregulation.[1] Exisulind thus stands as an unapproved but important investigational agent, illustrating the complex interplay between targeted efficacy, therapeutic index, and the potential for mechanism-based drug repurposing.
Exisulind is an investigational small molecule drug that represents the prototype of a class known as Selective Apoptotic Anti-neoplastic Drugs (SAANDs).[2] Its development was pioneered by Cell Pathways, which was later acquired by OSI Pharmaceuticals.[3] Chemically, Exisulind is the sulfone metabolite of the widely used NSAID, sulindac.[1] It is formed in the body through the irreversible oxidation of its parent compound.[17] This metabolic relationship is fundamental to its design, as the conversion to the sulfone form strips the molecule of the anti-inflammatory and COX-inhibitory activities associated with sulindac, thereby isolating its distinct anti-neoplastic properties.[4]
Throughout its long history of research and development, Exisulind has been referred to by numerous names and codes. Its non-proprietary name is Exisulind, while its most common chemical name is sulindac sulfone.[1] Tentative trade names included Aptosyn and Prevatac.[3] It was also identified by several research and development codes, most notably FGN-1 and CP 248.[1] Its unique identity is cataloged across major chemical and pharmacological databases through specific registry numbers, including its CAS Number, 59973-80-7, and its DrugBank ID, DB06246.[1]
Exisulind is a yellow crystalline powder.[19] Its chemical formula is $C_{20}H_{17}FO_{4}S$, with an average molecular weight of 372.41 g/mol and a monoisotopic mass of 372.083158364 Da.[2] The formal IUPAC name is 2-[(3Z)-6-fluoro-2-methyl-3-[(4-methylsulfonylphenyl)methylidene]inden-1-yl]acetic acid.[1] As a monocarboxylic acid, it is slightly acidic with a pKa of 4.33.[21] Its solubility profile shows that it is freely soluble in alcohol and polyethylene glycol but only slightly soluble in water.[20] This limited aqueous solubility is reflected in its calculated partition coefficient (XLogP3) of 3.5, indicating a lipophilic nature.[1]
Table 1: Chemical and Physical Identifiers of Exisulind
| Identifier Type | Value | Source(s) | 
|---|---|---|
| Generic Name | Exisulind | 1 | 
| Synonyms | Sulindac sulfone, Aptosyn, Prevatac, FGN-1 | 3 | 
| CAS Number | 59973-80-7 | 1 | 
| DrugBank ID | DB06246 | 2 | 
| IUPAC Name | 2-[(3Z)-6-fluoro-2-methyl-3-[(4-methylsulfonylphenyl)methylidene]inden-1-yl]acetic acid | 1 | 
| Molecular Formula | $C_{20}H_{17}FO_{4}S$ | 2 | 
| Molecular Weight | 372.41 g/mol (Average) | 2 | 
| SMILES | CC\1=C(C2=C(/C1=C\C3=CC=C(C=C3)S(=O)(=O)C)C=CC(=C2)F)CC(=O)O | 1 | 
| InChIKey | MVGSNCBCUWPVDA-MFOYZWKCSA-N | 1 | 
| Physical Appearance | Yellow crystalline powder / Yellow solid | 19 | 
| pKa | 4.33 (acidic) | 21 | 
| Solubility | Slightly soluble in water; freely soluble in alcohol | 20 | 
The central therapeutic concept behind Exisulind was to harness the observed anti-cancer properties of NSAIDs like sulindac while circumventing their dose-limiting toxicities.[4] Chronic NSAID use is associated with serious gastrointestinal and renal adverse events, which are directly attributable to the inhibition of cyclooxygenase (COX) enzymes and the subsequent disruption of prostaglandin synthesis.[8] Exisulind was specifically identified as the metabolite of sulindac that lacks meaningful COX-1 or COX-2 inhibitory activity.[1] Its ability to induce apoptosis is therefore independent of prostaglandin pathways and also operates separately from other common cancer pathways involving p53 or Bcl-2, establishing it as a new class of agent with a novel mechanism.[3]
The core of Exisulind's mechanism is the inhibition of cyclic guanosine monophosphate phosphodiesterases (cGMP-PDEs), a family of enzymes that degrade the second messenger cGMP.[2] Research has shown that Exisulind inhibits isoforms from the PDE2 and PDE5 gene families, which are frequently overexpressed in neoplastic tissues.[5] For instance, in human bladder tumor cells, Exisulind demonstrated inhibitory activity against PDE5 and PDE4 with 50% inhibitory concentrations ($IC_{50}$) of 112 µM and 116 µM, respectively.[7]
By inhibiting the enzymes responsible for cGMP breakdown, Exisulind treatment leads to a persistent and sustained elevation of intracellular cGMP levels.[5] This accumulation of cGMP is the critical initiating event in the drug's signaling cascade. Elevated cGMP, in turn, acts as an allosteric activator for cGMP-dependent protein kinase G (PKG).[2] The activation of PKG is the pivotal step that translates the upstream enzymatic inhibition into downstream cellular effects culminating in apoptosis.[20]
Once activated, PKG phosphorylates key cellular substrates that regulate cell survival and proliferation. Two primary downstream effects have been identified:
The therapeutic potential of Exisulind hinges on its ability to selectively target cancer cells while sparing normal tissues. This selectivity is believed to arise from the biological context of its target. The cGMP-PDE enzymes that Exisulind inhibits are significantly overexpressed in many precancerous and cancerous tissues compared to their normal counterparts.[2] This differential expression creates a therapeutic window: at clinically achievable concentrations, Exisulind can effectively inhibit the overabundant PDEs in tumor cells to trigger apoptosis, while the lower levels of the enzyme in normal cells are less affected, thus preserving their viability.[20]
In addition to its primary pro-apoptotic mechanism, Exisulind has been shown to exert other anti-cancer effects. Preclinical studies have provided evidence that it can inhibit angiogenesis, the formation of new blood vessels that tumors require to grow.[3] This effect may also be linked to the PKG pathway, which has been shown to down-regulate the expression of vascular endothelial growth factor (VEGF).[29] Furthermore, Exisulind is a potent inhibitor of aldose reductase (AKR1B1), an enzyme involved in the polyol pathway, with an $IC_{50}$ of 367 nM.[2] While primarily studied in the context of diabetic complications, this activity may contribute to its overall cellular effects.
Exisulind is formulated for oral administration.[12] While comprehensive absorption data for Exisulind itself is limited in the available literature, studies of more potent structural analogs provide insight into the class. For example, the related SAAND, CP-461, demonstrated rapid absorption, with maximum plasma concentrations ($T_{max}$) achieved between 1 to 2 hours after dosing.[32]
Specific data regarding the volume of distribution and plasma protein binding of Exisulind are not detailed in the provided materials. However, its parent drug, sulindac, is known to be highly protein-bound, a characteristic common to many NSAIDs and their derivatives.[33]
The metabolism of Exisulind is intrinsically linked to its origin. It is not a primary drug but rather an active metabolite of sulindac.[1] After oral administration, sulindac, which is a sulfoxide, undergoes extensive biotransformation in the body. This includes a reversible reduction to sulindac sulfide (the primary COX-inhibiting, anti-inflammatory metabolite) and an irreversible oxidation to sulindac sulfone (Exisulind), the COX-sparing, pro-apoptotic agent.[4] The complex interplay between these metabolic pathways, which may include enterohepatic recirculation, complicates the overall pharmacokinetic profile.[34]
The primary route of elimination for Exisulind appears to be renal. Following administration of sulindac, approximately half of the dose is excreted in the urine, largely in the form of conjugated Exisulind.[1] Clinical studies in patients with FAP determined the serum half-life ($t_{1/2}$) of Exisulind to be in the range of 6 to 9 hours.[12] This pharmacokinetic parameter is consistent with the twice-daily (BID) dosing schedules used in the majority of its clinical trials and suggests that little drug accumulation occurs with repeated dosing.[12]
The development of Exisulind appears to have been pursued without a complete, early-stage characterization of its absorption, distribution, metabolism, and excretion (ADME) profile. A comprehensive population pharmacokinetic model for sulindac and its metabolites was only described much later, underscoring the complexity of its disposition.[34] This lack of a complete initial understanding of Exisulind's own pharmacokinetic behavior may have posed a challenge during its clinical development. Given that its dose-limiting toxicity was hepatic and that a relationship between plasma concentration and adverse events was observed, an incomplete grasp of its ADME properties could have made it difficult to precisely model the therapeutic window and effectively manage the risk of hepatotoxicity in large-scale trials.[6]
Exisulind was the subject of a broad and ambitious clinical development program, exploring its utility in the prevention and treatment of multiple cancers. The outcomes of these trials varied significantly by indication, painting a complex picture of its therapeutic potential.
The initial and most promising application for Exisulind was in the prevention of colorectal cancer, focusing on patients with hereditary and sporadic precancerous polyps.
This was the lead indication for Exisulind. A Phase I trial in FAP patients successfully established a maximum tolerated dose (MTD) of 300 mg twice daily. The dose-limiting toxicity was reversible hepatic dysfunction, which occurred at a dose of 400 mg twice daily.[12] Subsequent Phase II and III trials provided strong evidence of efficacy. A one-year extension of a pivotal Phase III trial demonstrated that Exisulind significantly reduced the formation of new polyps in FAP patients.[3] These positive findings were the cornerstone of the New Drug Application (NDA) submitted to the FDA.[3]
Following the success in FAP, Exisulind was evaluated for the regression of sporadic adenomas in the general population. A large, multicenter, placebo-controlled Phase III trial was completed.[3] A 12-month dose-response study within this program found that a daily dose of 400 mg Exisulind led to a statistically significant regression of sporadic adenomas compared to placebo.[8] However, this efficacy was accompanied by a higher incidence of toxicity, particularly elevated liver enzymes and abdominal pain, foreshadowing the safety concerns that would ultimately derail its approval.[36]
Exisulind was extensively studied in various stages of prostate cancer, from preventing recurrence to treating advanced disease.
A key randomized, placebo-controlled trial investigated Exisulind (250 mg twice daily for 12 months) in 96 men who had rising PSA levels after a radical prostatectomy, indicating biochemical recurrence.[9] The results were statistically significant: Exisulind suppressed the rate of PSA increase compared to placebo, with a particularly strong effect in patients at high risk for developing metastatic disease. Furthermore, it significantly lengthened the PSA doubling time in this high-risk group.[9] The drug was reported to be well tolerated in this study.[9]
Several Phase II trials, including NCT00078910 and NCT00166478, were conducted to assess the biological activity of Exisulind when given as a neoadjuvant treatment for approximately four weeks before a planned radical prostatectomy.[16] While one study confirmed that the drug was well-tolerated with minimal side effects, it failed to demonstrate a significant increase in apoptosis in the prostate tissue at the dose and duration tested, suggesting limited preoperative activity.[39]
Based on strong preclinical data showing tumor growth inhibition and synergistic activity with chemotherapy, particularly taxanes, a large program of clinical trials was initiated in lung cancer.[6] Exisulind was tested in combination with a wide array of standard-of-care agents, including docetaxel, paclitaxel, carboplatin, vinorelbine, gemcitabine, and etoposide.[3]
Despite the compelling preclinical rationale, the clinical outcomes in lung cancer were uniformly negative. This disconnect between laboratory findings and patient outcomes represents a significant challenge in oncology drug development. The strong evidence of synergy in animal models, which justified the extensive and costly clinical program, did not translate into a tangible benefit for patients. A Phase II study combining Exisulind with weekly docetaxel for second-line treatment of NSCLC reported no objective responses and was terminated for futility.[10] Similarly, a large Phase I/II trial of Exisulind with a docetaxel and carboplatin combination in metastatic NSCLC found that adding Exisulind did not improve the response rate, duration of response, or overall survival compared to historical data for the chemotherapy doublet alone.[11] The investigators concluded that further development of Exisulind in this setting was not warranted.[11] This failure may be attributable to a number of factors, including species-specific differences in pharmacology, an inability to achieve synergistic drug concentrations in human tumors without causing unacceptable toxicity, or complexities of the human tumor microenvironment not captured by preclinical models.
Table 2: Summary of Key Clinical Trials for Exisulind
| Indication | Trial Phase | Reference / NCT ID | Intervention | Primary Outcome(s) | Key Result Summary | 
|---|---|---|---|---|---|
| Familial Adenomatous Polyposis (FAP) | Phase I | van Stolk R, et al. 2000 | Exisulind 200-400 mg BID | MTD, Safety | MTD established at 300 mg BID; DLT was reversible hepatotoxicity. 12 | 
| FAP | Phase III (Extension) | Cell Pathways Announcement | Exisulind | New polyp formation | Significantly reduced new polyp formation over 24 months. 3 | 
| Sporadic Colonic Polyps | Phase III | Arber N, et al. 2005 | Exisulind 400 mg/day vs. Placebo | Polyp size regression | Significant regression of adenomas, but associated with increased toxicity. 8 | 
| Prostate Cancer (Biochemical Recurrence) | Phase II/III | Goluboff ET, et al. 2001 | Exisulind 250 mg BID vs. Placebo | Change in PSA | Significantly suppressed PSA increase and lengthened PSA doubling time. 9 | 
| Prostate Cancer (Neoadjuvant) | Phase II | NCT00078910 | Exisulind prior to prostatectomy | Apoptosis in tumor tissue | Well tolerated but no significant effect on apoptotic biomarkers. 38 | 
| NSCLC (2nd Line) | Phase II | Hanna N, et al. 2007 | Exisulind + Docetaxel | Objective Response Rate (ORR) | No objective responses; trial deemed inactive. 10 | 
| NSCLC (1st Line, Metastatic) | Phase I/II | Treat J, et al. 2005 | Exisulind + Docetaxel/Carboplatin | ORR, Survival | ORR 23%; no apparent enhancement of activity or survival vs. historical controls. 11 | 
Across multiple clinical trials, Exisulind was often described as being generally well tolerated, with most adverse events being mild to moderate in severity and reversible upon dose reduction or discontinuation.[9] This was particularly noted in studies involving patients with prostate cancer, where the safety profile appeared favorable.[9]
The most consistently reported side effects were gastrointestinal in nature. These included dyspepsia (indigestion), nausea, vomiting, and abdominal pain.[6] Other common non-hematologic toxicities included fatigue, anorexia, and headache.[11]
The critical safety issue that defined Exisulind's clinical trajectory and ultimately prevented its approval was dose-limiting hepatotoxicity.[13] In clinical trials, reversible and typically asymptomatic elevations in liver function tests (transaminases) were consistently identified as the primary dose-limiting toxicity.[6] The Phase I study in FAP patients clearly established this, where four of six patients treated at the 400 mg twice-daily dose level experienced this effect, leading to the establishment of the MTD at 300 mg twice daily.[12]
This created a significant challenge for its development as a chemopreventive agent. A primary goal in designing Exisulind was to create a safer alternative to its parent NSAID, sulindac, by eliminating the COX-mediated gastrointestinal toxicities.[8] While this objective was largely met, the molecule's own intrinsic hepatotoxicity emerged as a new, insurmountable barrier. The dose required to achieve a consistent and statistically significant reduction in colorectal polyps was found to be too close to the dose that caused an unacceptable level of liver toxicity.[13] For a drug intended for long-term use in a preventative setting, where patients are otherwise healthy, the tolerance for potential organ damage is extremely low. This unfavorable therapeutic index made the overall risk-benefit assessment untenable for the FAP indication.
Pharmacological data indicate several potential drug-drug interactions for Exisulind:
Exisulind's journey with the U.S. Food and Drug Administration (FDA) was focused on the FAP indication and was marked by early promise followed by a definitive regulatory setback.
Years after its development for cancer was largely abandoned, Exisulind found a potential new life through a regulatory pathway in Europe for a different disease.
Exisulind remains an investigational drug. It has not received marketing authorization from any major regulatory body worldwide for any clinical indication. Its primary value today lies in its role as a pioneering SAAND and a chemical probe for studying the cGMP-PKG signaling pathway. It continues to serve as a lead compound for the design of next-generation PDE inhibitors with potentially improved safety profiles.[29]
The clinical story of Exisulind is a clear illustration of the critical importance of the therapeutic index in drug development. For its primary chemoprevention indications—FAP and sporadic polyps—the drug faced an insurmountable risk-benefit challenge. The dose required to achieve consistent and meaningful polyp regression was inseparable from the dose that produced an unacceptable rate of hepatotoxicity.[8] In a preventative setting, where therapy is administered long-term to individuals who are not yet diagnosed with invasive cancer, the tolerance for significant organ toxicity is exceptionally low. The benefit of delaying or preventing polyps could not outweigh the risk of liver damage, leading to the unfavorable regulatory outcome.
The extensive investigation of Exisulind provides several key takeaways for pharmaceutical research:
The EMA's orphan designation for FCCM represents a scientifically astute attempt to repurpose Exisulind by leveraging its known mechanism against a new disease with a shared molecular pathology.[15] This move also highlights a crucial strategic consideration in drug development: the context of the disease profoundly alters the risk-benefit calculus. The FDA's rejection for FAP occurred in a setting where an effective, albeit invasive, preventative option (colectomy) exists, making the safety bar for a new drug extremely high. In contrast, FCCM is a life-threatening condition with no approved medical therapies; the only interventions are symptomatic management or high-risk neurosurgery.[15] In this scenario of high unmet medical need, the tolerance for a manageable side effect like reversible hepatotoxicity is substantially greater. The repurposing of Exisulind for FCCM strategically shifts the drug from a low-risk-tolerance setting to a high-risk-tolerance one, potentially creating a viable path to approval where one did not previously exist.
The legacy of Exisulind confirms the therapeutic potential of targeting the cGMP-PKG signaling pathway for cancer and other diseases. Future research should build on these lessons. The development of next-generation SAANDs or selective PDE inhibitors should be prioritized, with a focus on designing molecules that retain the on-target efficacy of Exisulind but have been structurally modified to mitigate or eliminate the chemical liabilities responsible for hepatotoxicity.[29] Furthermore, clinical investigation of Exisulind itself for the treatment of FCCM is warranted to determine if its mechanistic promise can translate into a meaningful clinical benefit for this underserved patient population.
Published at: October 27, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.