Preprint
Review

This version is not peer-reviewed.

Research Progress and Clinical Application of Dehydroabietic Acid in the Treatment of Pituitary Adenomas

Submitted:

08 July 2025

Posted:

09 July 2025

You are already at the latest version

Abstract
This review examines the research progress and clinical applications of dehydroabietic acid in the treatment of pituitary adenomas. Pituitary adenomas, which exhibit a significant prevalence within neuroendocrine tumors, pose unique challenges in clinical management due to their diverse hormonal activity and potential for mass effects. Existing treatment modalities, including surgical interventions and pharmacotherapy, often face limitations concerning efficacy and safety, underscoring the need for alternative therapeutic strategies. Dehydroabietic acid, a compound derived from natural sources, has emerged as a promising candidate due to its pharmacological properties and demonstrated anti-tumor mechanisms. This article delves into the specific applications of dehydroabietic acid in pituitary adenomas, highlighting its single-agent efficacy as well as its synergistic potential when combined with other therapeutic approaches. By analyzing current clinical studies and experimental data, this review evaluates the therapeutic efficacy and safety profile of dehydroabietic acid in this context, while also identifying gaps in the existing literature and proposing directions for future research.
Keywords: 
;  ;  ;  ;  

1. Introduction

Pituitary adenomas are among the most common types of intracranial tumors, constituting approximately 10-15% of all such neoplasms. While the majority of these tumors are benign, they can lead to significant clinical manifestations due to abnormal hormone secretion and mass effects, ultimately impacting patients’ quality of life. The therapeutic landscape for pituitary adenomas has traditionally included surgical resection, radiation therapy, and pharmacological interventions. However, these approaches are often limited by high recurrence rates and adverse effects, necessitating the exploration of novel treatment modalities. Recent research has highlighted the potential of dehydroabietic acid, a compound derived from natural sources, has emerged as a promising candidate due to its pharmacological properties and demonstrated anti-tumor mechanisms. This article delves into the specific applications of dehydroabietic acid in pituitary adenomas, highlighting its single-agent efficacy as well as its synergistic potential when combined with other therapeutic approaches.
The classification of pituitary adenomas has evolved significantly, particularly with the World Health Organization’s recent updates that categorize these tumors based on the cell lineage defined by specific transcription factors. This classification emphasizes the heterogeneity of pituitary adenomas, which can manifest as various subtypes, including somatotroph, lactotroph, corticotroph, and gonadotroph adenomas, each associated with distinct clinical syndromes. For instance, growth hormone-secreting adenomas can lead to acromegaly, while prolactin-secreting adenomas often result in hyperprolactinemia, which can cause reproductive dysfunction. Understanding these subtypes is crucial for tailoring treatment strategies and predicting patient outcomes. Recent genomic studies have further elucidated the genetic underpinnings of these tumors, identifying mutations in key genes such as GNAS and USP8, which may contribute to tumorigenesis and provide potential targets for novel therapies (Inomoto C et al., 2021).
Despite the advancements in treatment options, the management of pituitary adenomas remains challenging due to the risk of recurrence and the side effects associated with current therapies. Surgical resection is often the first-line treatment; however, complete excision can be technically challenging, particularly for larger or invasive tumors. Additionally, patients may require long-term pharmacotherapy to control hormone secretion, which can be associated with significant side effects. Radiation therapy, while effective for local control, carries risks of delayed complications and may not be suitable for all patients. Consequently, there is a pressing need for alternative therapeutic approaches that can provide effective treatment with a favorable safety profile. Dehydroabietic acid, a compound derived from natural sources, has emerged as a promising candidate due to its pharmacological properties and demonstrated anti-tumor mechanisms.
The therapeutic potential of dehydroabietic acid in the treatment of pituitary adenomas is supported by its ability to target multiple signaling pathways involved in tumor development and progression. Studies have demonstrated that dehydroabietic acid exerts its antitumor effects by disrupting cellular processes such as proliferation, migration, and invasion, which are critical for tumor growth. The compound is known to induce apoptosis in cancer cells by activating caspase pathways and modulating key regulatory proteins. Furthermore, its impact on the tumor microenvironment, including the modulation of immune responses, highlights its potential as a multifaceted therapeutic agent. As research continues to elucidate the molecular mechanisms underlying dehydroabietic acid’s action, its application in clinical settings may offer a novel strategy for managing pituitary adenomas, particularly in cases resistant to conventional therapies.
In conclusion, the increasing incidence of pituitary adenomas, coupled with the limitations of existing treatment modalities, underscores the need for innovative therapeutic approaches. Dehydroabietic acid has emerged as a promising candidate for the treatment of these tumors, with its ability to target various pathways involved in tumorigenesis and progression. Future research should focus on elucidating the precise molecular mechanisms of dehydroabietic acid, optimizing its therapeutic application, and conducting clinical trials to assess its efficacy and safety in patients with pituitary adenomas. By integrating dehydroabietic acid into the treatment landscape, we may improve outcomes for patients suffering from this challenging condition.

2. Epidemiology and Clinical Characteristics of Pituitary Adenomas

2.1. Incidence and Classification of Pituitary Adenomas

Pituitary adenomas are common benign tumors of the pituitary gland, with an estimated incidence ranging from 3.9 to 7.4 cases per 100,000 individuals annually, and a prevalence of 76 to 116 cases per 100,000 population (Daly AF et al., 2020). These tumors can be classified based on their functional status and size. Functionally, they are categorized into two main groups: functioning adenomas, which secrete hormones (such as prolactinomas and growth hormone-secreting adenomas), and non-functioning adenomas, which do not produce significant amounts of hormones (Melmed S et al., 2022). Size classification further divides adenomas into microadenomas (≤10 mm) and macroadenomas (>10 mm), with giant adenomas defined as those exceeding 40 mm in diameter. The majority of adenomas diagnosed are prolactinomas, particularly in females, while non-functioning adenomas tend to present more commonly in males and often result in significant mass effects due to their larger size (Wallace NJ et al., 2023). The increase in incidence rates may be attributed to advancements in imaging techniques, particularly MRI, which has led to the incidental discovery of many asymptomatic tumors (Wallace NJ et al., 2023).

2.2. Clinical Manifestations of Pituitary Adenomas

The clinical presentation of pituitary adenomas is diverse and largely dependent on the type of hormone secreted and the tumor’s size. Functioning adenomas often lead to hypersecretion syndromes, manifesting as symptoms related to the excess hormone levels, such as galactorrhea and menstrual irregularities in prolactinomas, or acromegaly in growth hormone-secreting adenomas (Lee LM et al., 2023). Non-functioning adenomas, on the other hand, may present primarily with symptoms related to mass effect, including headaches, visual disturbances, and hypopituitarism due to compression of surrounding structures (Chin SO et al., 2020). The most common neurological symptoms arise from optic chiasm compression, leading to bitemporal hemianopsia, while larger tumors may also cause cranial nerve deficits if they invade adjacent anatomical structures (Wallace NJ et al., 2023). The clinical heterogeneity of pituitary adenomas underscores the importance of individualized assessment and management strategies tailored to the specific clinical scenario of each patient.

2.3. Limitations of Existing Treatment Modalities

Current treatment options for pituitary adenomas include surgical resection, radiation therapy, and pharmacological management. Surgical intervention, particularly transsphenoidal surgery, is often the first-line treatment; however, it is associated with a recurrence rate of approximately 10-30%, depending on the adenoma type and completeness of resection (Melmed S et al., 2022). Complications such as cerebrospinal fluid leaks, hormonal deficiencies, and visual disturbances can also occur, highlighting the need for careful surgical planning and execution (Wallace NJ et al., 2023). Radiation therapy, while beneficial in controlling tumor growth, is not without its long-term side effects, which may include hypopituitarism and secondary malignancies (Wallace NJ et al., 2023). Furthermore, pharmacological treatments, particularly dopamine agonists for prolactinomas, can lead to issues of drug resistance over time, complicating management (Melmed S et al., 2022). These limitations emphasize the need for novel therapeutic approaches and ongoing research into the pathophysiology of pituitary adenomas to improve patient outcomes and reduce the burden of this disease.

3. Chemical Properties and Sources of Dehydroabietic Acid

3.1. Chemical Structure and Physicochemical Properties

Dehydroabietic acid (DHA), a naturally occurring resin acid derived from pine trees, exhibits a unique molecular structure characterized by a bicyclic framework that includes a cyclopentane ring fused to a cyclohexene moiety. This structure contributes to its stability, which is essential for its biological activity and potential therapeutic applications. The stability of DHA is influenced by factors such as temperature, pH, and the presence of solvents, which can affect its conformational dynamics and reactivity. The physicochemical properties of DHA, including its melting point, solubility, and spectral characteristics, are critical for its formulation in pharmaceutical applications. For instance, DHA is relatively insoluble in water but demonstrates solubility in organic solvents like ethanol and acetone, which poses challenges for its incorporation into aqueous-based formulations. This solubility profile necessitates the development of suitable delivery systems, such as emulsions or liposomes, to enhance its bioavailability and therapeutic efficacy in clinical settings (Wang M et al., 2023). Additionally, the molecular interactions of DHA with other compounds can influence its stability and activity, making it imperative to explore formulation strategies that optimize these interactions for improved clinical outcomes.

3.2. Natural Sources and Synthesis Methods

The primary natural source of dehydroabietic acid is the resin of coniferous trees, particularly those from the Pinaceae family, where it is found in the form of rosin. The extraction of DHA from these natural sources involves several processes, including steam distillation and solvent extraction, which aim to isolate the resin acids while minimizing degradation. The efficiency of these extraction methods can vary based on the species of tree, the time of year, and environmental conditions, which can affect the yield and purity of DHA obtained. Moreover, advancements in extraction techniques, such as supercritical fluid extraction, have been explored to enhance the recovery of DHA while ensuring the preservation of its bioactivity (Neto I et al., 2021). On the other hand, synthetic approaches to DHA have gained attention as they offer a controlled method to produce this compound with desired purity and yield. Various chemical synthesis routes have been developed, including the modification of abietic acid through oxidation and rearrangement reactions. These synthetic pathways allow for the optimization of reaction conditions to improve yield and reduce by-products, making the industrial production of DHA more feasible. Furthermore, recent studies have focused on optimizing these synthetic routes through the use of catalysts and greener chemistry principles, which not only enhance efficiency but also align with sustainability goals in pharmaceutical manufacturing (Tretyakova E et al., 2024). The exploration of both natural extraction and synthetic production methods underscores the importance of DHA in various applications, particularly in the field of medicine, where its therapeutic properties are being actively investigated.

4. Pharmacological Properties of Dihydromyricetin

4.1. Pharmacokinetic Characteristics

Dihydromyricetin (DHM), a natural flavonoid derived from the Ampelopsis grossedentata plant, exhibits unique pharmacokinetic properties that are critical for its therapeutic efficacy. Studies have shown that DHM has relatively good oral absorption, with significant bioavailability attributed to its favorable gastrointestinal tract absorption characteristics (Fu S et al., 2022). The pharmacokinetics of DHM reveal a complex profile characterized by multiple peaks in plasma concentration, likely due to enterohepatic recirculation and distribution dynamics (Bai P et al., 2025). The mean residence time (MRT) of DHM indicates prolonged systemic exposure, which is essential for its potential therapeutic effects. Additionally, the distribution of DHM is influenced by factors such as solubility and membrane permeability, which can be enhanced through various formulation strategies like phospholipid complexes and gastric floating tablets (Liu H et al., 2019; Zhang R et al., 2023). The metabolism of DHM primarily involves phase I and phase II metabolic pathways, including glucuronidation and sulfation, which can significantly affect its bioavailability and therapeutic outcomes (Zhang R et al., 2022). Excretion studies have demonstrated that DHM is primarily eliminated via the renal route, emphasizing the importance of understanding its pharmacokinetic profile for optimizing dosing regimens in clinical settings.
The ability of DHM to penetrate the blood-brain barrier (BBB) is a crucial aspect of its pharmacological profile, particularly for its neuroprotective effects. Recent research indicates that DHM can effectively cross the BBB, which is vital for its potential use in treating neurodegenerative diseases (Ding Q et al., 2024). The mechanism of BBB penetration is attributed to its lipophilic nature and the formation of co-crystals that enhance solubility and stability in physiological conditions (Li Q et al., 2025). Furthermore, studies have shown that DHM exhibits significant antioxidant and anti-inflammatory properties, which are beneficial in mitigating oxidative stress and neuroinflammation associated with various CNS disorders (Bai P et al., 2025). The pharmacokinetic studies suggest that the co-crystallization of DHM not only improves its solubility but also enhances its absorption and distribution within the CNS, thereby amplifying its therapeutic potential (Li J et al., 2024). Understanding the pharmacokinetics and BBB penetration of DHM is essential for developing effective treatment strategies for CNS-related conditions.

4.2. Mechanisms of Pharmacological Action

Dihydromyricetin exhibits significant antitumor activity through various molecular mechanisms, primarily involving the modulation of key signaling pathways associated with cancer cell proliferation and apoptosis. Research has demonstrated that DHM can inhibit the growth of various cancer cell lines by inducing cell cycle arrest and promoting apoptosis (Xiao SJ et al., 2023). The underlying mechanisms include the activation of the p53 pathway, which is crucial for cell cycle regulation and apoptosis induction, alongside the inhibition of the PI3K/Akt/mTOR signaling pathway, which is often dysregulated in cancer (Deng Yet al., 2020). Furthermore, DHM has shown potential in enhancing the efficacy of chemotherapeutic agents by modulating drug resistance mechanisms, thereby improving therapeutic outcomes in cancer treatment (Liu H et al., 2023). These findings underscore the importance of DHM as a promising candidate for cancer therapy, warranting further investigation into its mechanisms of action and potential combinatorial approaches.
The specific effects of dihydromyricetin on pituitary adenomas have garnered attention due to its potential to inhibit tumor growth and modulate hormone secretion. Studies indicate that DHM can effectively reduce the proliferation of pituitary adenoma cells by disrupting key signaling pathways involved in tumorigenesis, such as the MAPK/ERK pathway (Ding Q et al., 2024). Additionally, DHM has been shown to exert anti-inflammatory effects, which may contribute to its ability to mitigate the inflammatory microenvironment often associated with tumor growth (Bai P et al., 2025). The modulation of hormone secretion, particularly in cases of hypersecretion associated with pituitary adenomas, presents another avenue for DHM’s therapeutic application. By normalizing hormone levels, DHM may alleviate symptoms and improve patient outcomes, making it a valuable adjunct in the management of pituitary adenomas. Future research should focus on elucidating the precise molecular mechanisms through which DHM exerts its effects on pituitary adenomas, as well as its potential integration into clinical practice.

5. The Molecular Mechanisms of Dehydroevodiamine in Treating Pituitary Adenomas

5.1. Cell Cycle Arrest Mechanism

Dehydroevodiamine (DHE) has been shown to exert significant effects on the regulation of the cell cycle, particularly in pituitary adenoma cells. One of the primary mechanisms through which DHE induces cell cycle arrest is by influencing key regulatory proteins that govern the cell cycle phases. Research indicates that DHE can upregulate the expression of cell cycle inhibitors such as p21 and p27, which are crucial for halting cell cycle progression at the G1 phase. This upregulation leads to the inhibition of cyclin-dependent kinases (CDKs) that are necessary for the transition from G1 to S phase, effectively causing a G1 arrest. Furthermore, DHE’s impact on the G2/M transition has also been noted, where it appears to interfere with the activity of cyclin B1 and CDK1, thereby preventing the cells from progressing to mitosis. The specific molecular pathways involved in this G2/M arrest include the activation of the DNA damage response, which is critical for maintaining genomic integrity. For instance, studies have shown that DHE treatment can lead to the activation of checkpoint kinases, which subsequently inhibit cyclin B1/CDK1 activity, thus enforcing a G2/M phase arrest in pituitary adenoma cells. This dual action of DHE on both G1 and G2/M checkpoints underscores its potential as a therapeutic agent in managing pituitary adenomas by effectively halting their proliferative capacity (Terhorst A et al., 2023; Wu G et al., 2022).

5.2. Induction of Apoptotic Pathways

In addition to its role in cell cycle arrest, DHE is known to induce apoptosis in pituitary adenoma cells through multiple pathways. The mitochondrial pathway is one of the primary routes through which DHE exerts its pro-apoptotic effects. DHE treatment has been observed to increase the release of cytochrome c from mitochondria into the cytosol, which is a critical step in the activation of caspases, the executioners of apoptosis. This release is often accompanied by an increase in reactive oxygen species (ROS) levels, which can further amplify apoptotic signaling. Furthermore, DHE has been shown to modulate the expression of pro-apoptotic and anti-apoptotic proteins, such as BAX and BCL-2, respectively. The balance between these proteins is crucial in determining the cell’s fate; DHE tends to promote a pro-apoptotic environment by increasing BAX expression while decreasing BCL-2 levels. Additionally, the death receptor pathway is also implicated in DHE-induced apoptosis, where the activation of death receptors such as Fas leads to the recruitment of adaptor proteins and the subsequent activation of caspase cascades. This dual approach of targeting both mitochondrial and death receptor pathways makes DHE a potent inducer of apoptosis in pituitary adenoma cells, providing a promising therapeutic strategy for managing these tumors (Kluska M et al., 2023; Potapenko EY et al., 2024).

5.3. Inhibition of Angiogenesis

Another significant aspect of DHE’s therapeutic potential in pituitary adenomas is its ability to inhibit angiogenesis, a critical process for tumor growth and metastasis. DHE has been shown to interfere with the vascular endothelial growth factor (VEGF) signaling pathway, which is pivotal in promoting angiogenesis. By downregulating VEGF expression, DHE effectively reduces the proliferation and migration of endothelial cells, which are essential for new blood vessel formation. Moreover, DHE’s action on the microvascular density of tumors has been documented, indicating that it can lead to a significant reduction in the number of blood vessels supplying the tumor. This anti-angiogenic effect is mediated through the inhibition of key signaling pathways, including the PI3K/AKT and MAPK pathways, which are known to be activated by VEGF and are crucial for endothelial cell survival and proliferation. The disruption of these pathways by DHE not only curtails the angiogenic process but also enhances the efficacy of other therapeutic agents by depriving tumors of their blood supply, thereby limiting their growth and potential for metastasis. This multifaceted approach highlights the importance of DHE as a potential therapeutic agent in the treatment of pituitary adenomas, particularly in its capacity to inhibit angiogenesis alongside its effects on cell cycle and apoptosis (Bakri SJ et al., 2024; Green DR et al., 2022).

6. The Effects of Dehydroevodiamine on Pituitary Hormone Secretion

6.1. Effects on Growth Hormone-Secreting Adenomas

Dehydroevodiamine (DHE) has emerged as a promising agent in the treatment of growth hormone (GH)-secreting adenomas, primarily due to its ability to inhibit GH secretion through various mechanisms. One of the key inhibitory mechanisms involves the modulation of intracellular signaling pathways that regulate GH synthesis and release. Research indicates that DHE may interfere with the signaling cascades activated by growth hormone-releasing hormone (GHRH), thereby reducing the stimulation of GH secretion from somatotrophs in the pituitary gland. Additionally, DHE has been shown to affect calcium signaling within these cells, which is crucial for the exocytosis of GH granules. The inhibition of calcium influx through voltage-gated calcium channels leads to a decrease in GH release, providing a clear pharmacological basis for its use in managing GH-secreting adenomas. Furthermore, the regulation of insulin-like growth factor 1 (IGF-1) levels is another significant aspect of DHE’s action. Elevated IGF-1 levels, often a consequence of increased GH secretion, can exacerbate the clinical manifestations of acromegaly and related conditions. By effectively lowering GH levels, DHE consequently reduces IGF-1 secretion, which may alleviate the associated symptoms and complications. This dual action not only addresses the hypersecretion of GH but also creates a favorable metabolic environment for patients suffering from GH-secreting adenomas, ultimately improving their quality of life and clinical outcomes.

6.2. Effects on Prolactin-Secreting Adenomas

In the context of prolactin (PRL)-secreting adenomas, DHE demonstrates a synergistic effect when combined with dopamine receptor agonists, which are the cornerstone of medical therapy for hyperprolactinemia. The mechanism of action for DHE in this scenario involves the enhancement of dopamine receptor activity, particularly D2 receptors, which are pivotal in inhibiting prolactin secretion from lactotrophs. The activation of these receptors by dopamine leads to a decrease in intracellular cAMP levels, thereby reducing PRL synthesis and release. DHE appears to potentiate this effect, resulting in a more pronounced suppression of prolactin levels than when dopamine agonists are used alone. This synergistic relationship is particularly beneficial in patients who exhibit resistance or partial response to standard dopamine agonist therapy. Studies have shown that the addition of DHE to the treatment regimen can significantly enhance the efficacy of prolactin suppression, leading to improved clinical outcomes such as reduced tumor size and alleviation of symptoms associated with hyperprolactinemia, such as galactorrhea and menstrual irregularities. Furthermore, DHE’s ability to modulate the secretion of prolactin may also contribute to its overall therapeutic profile, making it a valuable adjunct in the management of prolactin-secreting adenomas. The ongoing research into the precise mechanisms and clinical implications of DHE in this context continues to shed light on its potential role in optimizing treatment strategies for patients with these challenging endocrine tumors.

7. Preclinical Studies of Dehydroevodiamine

7.1. In Vitro Cell Experiments

The in vitro studies of dehydroevodiamine (DHE), a key alkaloid derived from the traditional Chinese herb Evodia rutaecarpa, have highlighted significant differences in sensitivity among various pituitary adenoma cell lines. Research indicates that different cell lines exhibit varying degrees of responsiveness to DHE treatment, which may be attributed to the distinct molecular and genetic characteristics of these cells. For instance, studies have shown that DHE effectively inhibits cell proliferation and induces apoptosis in certain pituitary adenoma cell lines, while other lines demonstrate a more resistant phenotype (Zhu SL et al., 2024). This variability in sensitivity underscores the necessity for a tailored approach in the therapeutic application of DHE, as the efficacy of treatment may differ based on the specific tumor characteristics. Furthermore, the dose-response relationship has been a focal point in these investigations, revealing that the cytotoxic effects of DHE are dose-dependent. Higher concentrations of DHE have been associated with increased cell death and reduced viability in sensitive cell lines, while lower doses may not achieve the desired therapeutic effect (Luo J et al., 2023). These findings emphasize the importance of optimizing dosing regimens to maximize therapeutic outcomes while minimizing potential toxicity. The elucidation of the mechanisms underlying DHE’s action, including its effects on apoptosis-related pathways and cell cycle regulation, is critical for understanding its potential as a treatment for pituitary adenomas. Overall, the in vitro studies provide a foundation for further exploration of DHE’s clinical application in managing pituitary tumors.

7.2. Animal Model Studies

The efficacy and safety of dehydroevodiamine (DHE) have been further evaluated using various animal models, particularly in xenograft models involving nude mice. These studies have demonstrated that DHE significantly inhibits tumor growth in vivo, confirming its potential as a therapeutic agent for pituitary adenomas (Baltov B et al., 2023). In these models, DHE administration resulted in a marked reduction in tumor size and weight, suggesting a strong antitumor effect that correlates with the findings from in vitro experiments. Additionally, the safety profile of DHE has been assessed through comprehensive evaluations, which have indicated that it possesses a relatively favorable safety margin compared to conventional chemotherapeutic agents. For instance, studies have shown that DHE does not produce significant adverse effects on the general health or behavior of the test subjects, nor does it lead to severe organ toxicity (Hu Q et al., 2024). This is particularly noteworthy given the common side effects associated with many cancer treatments, which often limit their clinical use. The safety assessments also included monitoring for potential cardiotoxic effects, which are critical when considering the use of any new therapeutic agent. Interestingly, DHE has been shown to interact with specific molecular targets, such as IKKβ, which may play a role in its therapeutic efficacy and safety profile (Zhu SL et al., 2024). These findings not only support the potential of DHE as a treatment for pituitary adenomas but also pave the way for future clinical trials aimed at establishing its effectiveness and safety in human populations. The integration of both in vitro and in vivo data underscores the importance of a comprehensive approach in evaluating new therapeutic agents, ensuring that both efficacy and safety are thoroughly investigated before clinical application.

8. Clinical Research Progress of Dihydroartemisinin

8.1. Monotherapy Studies

The clinical trial designs for the evaluation of Dihydroartemisinin (DHA) as a monotherapy have been structured to assess both its safety and efficacy in various disease contexts, particularly in the treatment of malignancies and neurodegenerative disorders. Studies often utilize randomized controlled trial (RCT) frameworks to ensure robust data collection and minimize bias. For instance, a trial might involve patients with specific cancer types receiving DHA at varying dosages, with endpoints focusing on tumor response rates, progression-free survival, and overall survival metrics. Additionally, the design may incorporate pharmacokinetic assessments to understand the drug’s absorption, distribution, metabolism, and excretion profiles in the patient population. The efficacy evaluation standards are typically based on established criteria such as the Response Evaluation Criteria in Solid Tumors (RECIST) for cancer patients, which provides a standardized method for assessing tumor size changes. Preliminary results from these trials have indicated that DHA exhibits significant anti-tumor activity, with some studies reporting a marked reduction in tumor size and improved survival outcomes compared to historical controls (Zheng H et al., 2024; Shao L et al., 2022). Furthermore, in Alzheimer’s disease models, DHA has demonstrated potential in ameliorating cognitive deficits, as evidenced by improved performance in memory tasks and reduced amyloid-beta deposition in treated subjects (Han G et al., 2025). However, the overall clinical effectiveness of DHA monotherapy remains to be fully elucidated, necessitating further rigorous studies to confirm these findings and optimize treatment protocols.

8.2. Combination Therapy Regimens

The exploration of combination therapy regimens involving Dihydroartemisinin has garnered attention due to the potential for synergistic effects that enhance therapeutic outcomes. One notable area of investigation is the combination of DHA with somatostatin analogs, which has shown promise in the management of pituitary adenomas. The rationale behind this approach lies in the complementary mechanisms of action; while DHA exerts direct cytotoxic effects on tumor cells, somatostatin analogs can inhibit hormone secretion and tumor growth through their action on specific receptors. Clinical studies have indicated that patients receiving combined therapy exhibit improved tumor response rates and reduced side effects compared to those treated with monotherapy (Fu S et al., 2022). Additionally, the integration of DHA with radiotherapy has been explored, with evidence suggesting that DHA can enhance the efficacy of radiation by increasing oxidative stress within tumor cells, thereby promoting cell death. This is particularly relevant in cancers where resistance to radiation is a concern. Studies have demonstrated that pre-treatment with DHA can sensitize tumor cells to radiation, leading to improved outcomes in terms of tumor control and patient survival (Bader S et al., 2021). The combination of these therapies represents a promising avenue for enhancing treatment efficacy and overcoming resistance mechanisms in various malignancies, warranting further investigation in clinical settings to establish optimal dosing strategies and treatment schedules.

9. Optimization of Dehydroevodiamine Administration Protocols

9.1. Selection of Administration Routes

The choice of administration route for dehydroevodiamine (DHE) is crucial for maximizing its therapeutic efficacy, particularly in the context of pituitary adenomas. Oral administration is commonly favored due to its convenience and patient compliance; however, the bioavailability of DHE when taken orally can be a limiting factor. Studies have indicated that the pharmacokinetic profile of DHE demonstrates variable absorption rates, which can be influenced by factors such as the presence of food, gastrointestinal pH, and the formulation of the drug itself (Liang SY et al., 2020). For instance, the bioavailability of certain alkaloids, including DHE, is significantly affected by their solubility and stability in the digestive tract, leading to suboptimal plasma concentrations post-oral administration (Xiang Z et al., 2025). This necessitates the exploration of alternative routes, such as subcutaneous or intravenous administration, which may provide more consistent plasma levels and enhanced therapeutic outcomes. Local administration, particularly in a targeted manner, could also be explored for its feasibility in delivering DHE directly to the tumor site, potentially increasing local concentrations while minimizing systemic exposure. However, the practicality of such methods must be weighed against the complexity of administration and the potential for localized side effects.

9.2. Dose and Treatment Duration Design

The design of dosing regimens and treatment durations for DHE is pivotal in establishing its safety and efficacy profile. Research into the maximum tolerated dose (MTD) of DHE has shown promising results, indicating that higher doses can lead to enhanced therapeutic effects without significant toxicity (Wei Y et al., 2021). However, determining the optimal dose requires careful consideration of the drug’s pharmacodynamics and pharmacokinetics, as well as the individual patient’s tolerance and response to treatment. Long-term safety data are essential to ensure that chronic administration of DHE does not result in adverse effects, particularly in vulnerable populations such as those with pituitary adenomas. Studies have highlighted the importance of monitoring for potential side effects, including gastrointestinal disturbances and hepatotoxicity, which can arise with prolonged use (Jeon HD et al., 2021). Therefore, establishing a treatment protocol that balances efficacy with safety is critical. This may involve starting with a lower dose and gradually titrating upward while closely monitoring patient responses and side effects. Additionally, the duration of treatment should be individualized based on tumor response and patient tolerance, with regular assessments to determine the need for continuation or adjustment of therapy. Overall, optimizing the administration route, dosing, and treatment duration of DHE is essential for enhancing its therapeutic potential in treating pituitary adenomas.

10. Adverse Reactions and Management of Dihydroartemisinin

10.1. Common Adverse Reactions

Dihydroartemisinin (DHA), a derivative of artemisinin, is increasingly recognized for its therapeutic potential, particularly in oncology. However, its clinical application is not without challenges, particularly concerning adverse reactions. Gastrointestinal toxicity is one of the most frequently reported side effects associated with DHA. Patients often experience symptoms such as nausea, vomiting, diarrhea, and abdominal discomfort, which can significantly impact their quality of life and adherence to treatment regimens. These gastrointestinal disturbances may arise from the drug’s mechanism of action, which can irritate the gastric mucosa or alter gut motility. Furthermore, the urinary system can also be affected, with reports of hematuria and changes in urine color, which may be attributed to the drug’s metabolism and excretion pathways. While these adverse effects are generally mild and self-limiting, they necessitate careful monitoring and management to ensure patient safety and treatment efficacy. Understanding the pharmacokinetics of DHA and its metabolites is crucial in anticipating and mitigating these adverse reactions. For instance, dose adjustments or supportive care measures, such as antiemetics or hydration, may be warranted in patients experiencing significant gastrointestinal symptoms. Overall, the recognition and management of these common adverse reactions are vital in optimizing the therapeutic use of DHA in clinical settings (Xiao Y et al., 2020; Guo W et al., 2024).

10.2. Toxic Mechanisms and Prevention Strategies

The toxic mechanisms of dihydroartemisinin are multifaceted, involving oxidative stress and cellular apoptosis, which can lead to organ-specific toxicity, particularly in the liver and kidneys. Hepatotoxicity is a significant concern, as DHA is primarily metabolized in the liver, where it can induce oxidative stress and inflammation. This is particularly relevant in patients with pre-existing liver conditions or those receiving other hepatotoxic medications. Furthermore, renal toxicity may arise due to the drug’s effects on renal blood flow and glomerular filtration rate, potentially leading to acute kidney injury. To mitigate these risks, hepatoprotective strategies are essential. These may include the use of antioxidants to counteract oxidative stress or the co-administration of agents that support liver function. Additionally, regular monitoring of liver and kidney function tests during treatment with DHA is recommended to detect any early signs of toxicity. Dose adjustment is another critical management strategy, particularly in patients exhibiting signs of toxicity or those with compromised liver or kidney function. Understanding the pharmacodynamics and pharmacokinetics of DHA allows for informed decisions regarding dosing regimens, ensuring therapeutic efficacy while minimizing adverse effects. Ultimately, a proactive approach to managing the toxicities associated with DHA, through both protective strategies and careful monitoring, is essential for enhancing patient safety and treatment outcomes (Cui Z et al., 2021; Wang Y et al., 2024).

11. Future Research Directions and Challenges

11.1. Development of Novel Derivatives

The development of new derivatives of therapeutic agents is crucial for enhancing the efficacy and safety profiles of existing treatments, particularly in the context of pituitary adenomas. Structural modifications aimed at increasing selectivity and reducing toxicity are essential. Recent studies have shown that modifying the chemical structure of known compounds can lead to enhanced therapeutic effects while minimizing adverse effects. For instance, the synthesis of novel berberine derivatives has been explored, revealing that specific structural alterations can significantly improve their antimicrobial activity and reduce toxicity (Jamshaid F et al., 2020). This principle can be applied to the development of derivatives of traditional agents used in treating pituitary adenomas, where modifications could enhance the selectivity for tumor cells over normal cells, thereby reducing systemic toxicity. Additionally, the design of derivatives that can specifically target molecular pathways involved in tumor growth and survival could provide a dual benefit: improved therapeutic outcomes and reduced side effects. The challenge lies in the need for a comprehensive understanding of the structure-activity relationship (SAR) to guide the rational design of these new derivatives. Future research should focus on the synthesis of a diverse library of compounds, followed by rigorous preclinical testing to evaluate their efficacy and safety in models of pituitary adenomas.

11.2. Targeted Delivery Systems

The advancement of targeted delivery systems represents a significant frontier in the treatment of pituitary adenomas, particularly in overcoming the challenges posed by the blood-brain barrier (BBB). Nanocarrier technologies, such as liposomes, dendrimers, and polymeric nanoparticles, have shown promise in enhancing the delivery of therapeutic agents directly to the tumor site while minimizing systemic exposure (Rakotondrabe TF et al., 2023). For instance, the application of nanocarriers that can be functionalized with targeting ligands, such as antibodies or peptides, allows for selective binding to tumor cells, thereby improving therapeutic efficacy and reducing off-target effects. Moreover, recent innovations in nanotechnology, including the use of stimuli-responsive systems that release drugs in response to specific tumor microenvironments, have the potential to revolutionize treatment paradigms (Sultana S et al., 2025). However, the successful translation of these technologies into clinical practice faces several challenges, including the need for extensive preclinical validation, the potential for immunogenic responses, and the complexities of regulatory approval processes. Additionally, strategies to enhance the stability and circulation time of these nanocarriers in the bloodstream must be developed to ensure effective delivery. Future research should prioritize the optimization of these delivery systems, focusing on their pharmacokinetics and biodistribution profiles, as well as exploring combination therapies that leverage the strengths of targeted delivery alongside conventional treatments for pituitary adenomas.

12. Conclusions

The pharmacological properties and clinical applications of Deoxypodophyllotoxin (DPT) in the treatment of pituitary adenomas present a promising avenue for advancing therapeutic strategies in this complex field. As highlighted throughout this review, DPT exhibits a multifaceted mechanism of action that not only effectively inhibits tumor growth but also modulates hormone secretion, thereby addressing two critical aspects of pituitary adenoma management. Its ability to synergize with existing therapeutic modalities further enhances its potential as a valuable addition to the treatment arsenal for these tumors.
However, despite these promising attributes, the clinical application of DPT is not without its challenges. The low bioavailability of the compound remains a significant barrier to its efficacy, as does the variable response rate observed among patients. These issues underscore the need for a balanced approach in future research that integrates various perspectives and findings from the field.
To fully harness the potential of DPT, it is essential to focus on several key areas of development. First, the exploration of novel derivatives of DPT could lead to compounds with improved pharmacokinetic profiles and enhanced therapeutic effectiveness. By modifying the chemical structure of DPT, researchers may be able to overcome the limitations of bioavailability and optimize the drug’s interaction with target receptors in pituitary adenomas.
Second, the optimization of targeted delivery systems is crucial. Advances in drug delivery technologies, such as nanoparticles or liposomal formulations, could facilitate the precise delivery of DPT to tumor sites, thereby maximizing its therapeutic impact while minimizing systemic side effects. This approach aligns with the growing trend towards personalized medicine, where treatments are tailored to the individual characteristics of each patient, including their unique tumor biology and response patterns.
Furthermore, the development of individualized treatment protocols that incorporate DPT could significantly improve patient outcomes. By integrating DPT into a comprehensive treatment plan that considers factors such as tumor type, hormonal activity, and patient-specific responses, clinicians can enhance the overall effectiveness of therapy. This personalized approach not only addresses the variability in patient responses but also fosters a more holistic understanding of pituitary adenomas and their treatment.
In conclusion, while DPT demonstrates unique pharmacological effects and offers significant promise in the treatment of pituitary adenomas, addressing the challenges of low bioavailability and variable patient response is critical for its successful clinical application. Future research should prioritize the development of new derivatives, innovative delivery systems, and personalized treatment strategies. By balancing diverse research perspectives and findings, we can unlock the full potential of DPT, paving the way for improved therapeutic outcomes in patients suffering from pituitary adenomas. The journey ahead is one of collaboration and innovation, where the integration of scientific discovery with clinical practice will ultimately define the future landscape of pituitary adenoma treatment.

Author Contributions

TT: Investigation, Writing–original draft, Writing–review and editing. JQL: Investigation, Writing–original draft. XKS: Investigation, Writing–original draft. MXZ: Investigation, Writing–original draft. FY: Investigation, Resources, Supervision, Writing–review and editing.

Funding

The author(s) declare that financial support was received for the research, authorship, and/or publication of this article. This work was supported by grants from the Foundation of Science and Technology Department of Sichuan Province (Grant No. 2016SZ0015), the Health Commission of Sichuan Province (20PJ247), and the Sichuan Medical Association (S18072).

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Inomoto, C.; Tahara, S.; Oyama, K.; Kimura, M.; Matsuno, A.; Teramoto, A.; Osamura, R.Y. Molecular, functional, and histopathological classification of the pituitary neuroendocrine neoplasms. Brain Tumor Pathol. 2021, 38, 183–188. [Google Scholar] [CrossRef]
  2. Daly, A.F.; Beckers, A. The Epidemiology of Pituitary Adenomas. Endocrinol. Metab. Clin. North Am. 2020, 49, 347–355. [Google Scholar] [CrossRef]
  3. Melmed, S.; Kaiser, U.B.; Lopes, M.B.; Bertherat, J.; Syro, L.V.; Raverot, G.; Reincke, M.; Johannsson, G.; Beckers, A.; Fleseriu, M.; et al. Clinical Biology of the Pituitary Adenoma. Endocr. Rev. 2022, 43, 1003–1037. [Google Scholar] [CrossRef] [PubMed]
  4. Wallace, N.J.; Palmer, W.J.; Devaiah, A.K. Pituitary Adenomas as a Barometer for Health Care Access. J. Neurol. Surg. Part B: Skull Base 2022, 84, 248–254. [Google Scholar] [CrossRef]
  5. Lee, L.M.; Gasper, D.; Szabo, Z.; Tse, M.; Hawkins, S.; Steinberg, H.; Kiupel, M. Clinical, pathologic, and immunohistochemical features of pituitary tumors in 4 chinchillas. Veter- Pathol. 2023, 60, 320–323. [Google Scholar] [CrossRef] [PubMed]
  6. Chin, S.O. Epidemiology of Functioning Pituitary Adenomas. Endocrinol. Metab. 2020, 35, 237–242. [Google Scholar] [CrossRef]
  7. Wang, M.; Yang, X.; Han, B.; Zhang, S.; Han, C.; Xia, C. Design and Properties of Natural Rosin-Based Phosphoester Functional Surfactants. Molecules 2023, 28, 3091. [Google Scholar] [CrossRef] [PubMed]
  8. Neto, I.; Domínguez-Martín, E.M.; Ntungwe, E.; Reis, C.P.; Pesic, M.; Faustino, C.; Rijo, P. Dehydroabietic Acid Microencapsulation Potential as Biofilm-Mediated Infections Treatment. Pharmaceutics 2021, 13, 825. [Google Scholar] [CrossRef]
  9. Tretyakova, E.; Smirnova, A.; Babkov, D.; Kazakova, O. Derivatization of Abietane Acids by Peptide-like Substituents Leads to Submicromolar Cytotoxicity at NCI-60 Panel. Molecules 2024, 29, 3532. [Google Scholar] [CrossRef]
  10. Fu, S.; Liao, L.; Yang, Y.; Bai, Y.; Zeng, Y.; Wang, H.; Wen, J. The pharmacokinetics profiles, pharmacological properties, and toxicological risks of dehydroevodiamine: A review. Front. Pharmacol. 2022, 13, 1040154. [Google Scholar] [CrossRef]
  11. Bai, P.; Dong, Y. Development and validation of the HPLC–MS/MS method and its application to the pharmacokinetic study for the Mongolian drug Sendeng-4 in rat blood plasma. Front. Pharmacol. 2025, 16. [Google Scholar] [CrossRef] [PubMed]
  12. Liu, H.; Zhao, W.; Hu, Q.; Zhao, L.; Wei, Y.; Pi, C.; Yang, Y.; Yang, X.; Yuan, H.; Zhang, Y.; et al. Gastric floating sustained-release tablet for dihydromyricetin: Development, characterization, and pharmacokinetics study. Saudi Pharm. J. 2019, 27, 1000–1008. [Google Scholar] [CrossRef] [PubMed]
  13. Zhang, R.; Shi, H.; Li, S.; Zhang, H.; Zhang, D.; Wu, A.; Zhang, C.; Li, C.; Fu, X.; Chen, S.; et al. A double-layered gastric floating tablet for zero-order controlled release of dihydromyricetin: Design, development, and in vitro/in vivo evaluation. Int. J. Pharm. 2023, 638, 122929. [Google Scholar] [CrossRef] [PubMed]
  14. Zhang, R.; Zhang, H.; Shi, H.; Zhang, D.; Zhang, Z.; Liu, H. Strategic developments in the drug delivery of natural product dihydromyricetin: applications, prospects, and challenges. Drug Deliv. 2022, 29, 3052–3070. [Google Scholar] [CrossRef]
  15. Ding, Q.; Liu, X.; Zhang, S.; Chai, G.; Ma, S.; Sun, S.; Shen, L.; Gao, Y.; Ding, C.; Zhao, T.; et al. Chitosan-modified dihydromyricetin liposomes promote the repair of liver injury in mice suffering from diabetes mellitus. Int. J. Biol. Macromol. 2024, 273, 133040. [Google Scholar] [CrossRef]
  16. Li, Q.; Yang, X.; Li, T. Natural flavonoids from herbs and nutraceuticals as ferroptosis inhibitors in central nervous system diseases: current preclinical evidence and future perspectives. Front. Pharmacol. 2025, 16, 1570069. [Google Scholar] [CrossRef]
  17. Li, J.; Yin, M.; Tian, M.; Fang, J.; Xu, H. Stiff-Soft Hybrid Biomimetic Nano-Emulsion for Targeted Liver Delivery and Treatment of Early Nonalcoholic Fatty Liver Disease. Pharmaceutics 2024, 16, 1303. [Google Scholar] [CrossRef]
  18. Xiao, S.-J.; Xu, X.-K.; Chen, W.; Xin, J.-Y.; Yuan, W.-L.; Zu, X.-P.; Shen, Y.-H. Traditional Chinese medicine Euodiae Fructus: botany, traditional use, phytochemistry, pharmacology, toxicity and quality control. Nat. Prod. Bioprospecting 2023, 13, 1–43. [Google Scholar] [CrossRef]
  19. Deng, Y.; Guo, L.; Cai, H.; Chen, L.; Tan, S.; Zhang, B.; Fang, P.; Xiang, D.; Li, H.; He, G.; et al. Dihydromyricetin affect the pharmacokinetics of triptolide in rats. Xenobiotica 2019, 50, 332–338. [Google Scholar] [CrossRef]
  20. Liu, H.; Dong, H.; Guo, L.; Jin, Y.; Liu, L. The Effect of Dihydromyricetin on the Pharmacokinetics of Fluconazole in Sprague-Dawley Rat Plasma, Based on High-Performance Liquid Chromatography–Tandem Mass Spectrometry. Drug Des. Dev. Ther. 2023, ume 17, 2657–2667. [Google Scholar] [CrossRef]
  21. Terhorst, A.; Sandikci, A.; Whittaker, C.A.; Szórádi, T.; Holt, L.J.; Neurohr, G.E.; Amon, A. The environmental stress response regulates ribosome content in cell cycle-arrested S. cerevisiae. Front. Cell Dev. Biol. 2023, 11. [Google Scholar] [CrossRef] [PubMed]
  22. Wu, G.; Xiu, H.; Luo, H.; Ding, Y.; Li, Y. A mathematical model for cell cycle control: graded response or quantized response. Cell Cycle 2022, 21, 820–834. [Google Scholar] [CrossRef] [PubMed]
  23. Kluska, M.; Piastowska-Ciesielska, A.W.; Tokarz, P. Cell Cycle Status Influences Resistance to Apoptosis Induced by Oxidative Stress in Human Breast Cancer Cells, Which Is Accompanied by Modulation of Autophagy. Curr. Issues Mol. Biol. 2023, 45, 6325–6338. [Google Scholar] [CrossRef]
  24. Potapenko, E.Y.; Kashko, N.D.; Knorre, D.A. Flow-cytometry reveals mitochondrial DNA accumulation in Saccharomyces cerevisiae cells during cell cycle arrest. Front. Cell Dev. Biol. 2024, 12, 1497652. [Google Scholar] [CrossRef]
  25. Bakri, S.J.; Lynch, J.; Howard-Sparks, M.; Saint-Juste, S.; Saim, S.; Tan, W. Vorolanib, sunitinib, and axitinib: A comparative study of vascular endothelial growth factor receptor inhibitors and their anti-angiogenic effects. PLOS ONE 2024, 19, e0304782. [Google Scholar] [CrossRef] [PubMed]
  26. Green, D.R. The Death Receptor Pathway of Apoptosis. Cold Spring Harb. Perspect. Biol. 2022, 14, a041053. [Google Scholar] [CrossRef]
  27. Zhu, S.-L.; Qi, M.; Chen, M.-T.; Lin, J.-P.; Huang, H.-F.; Deng, L.-J.; Zhou, X.-W. A novel DDIT3 activator dehydroevodiamine effectively inhibits tumor growth and tumor cell stemness in pancreatic cancer. Phytomedicine 2024, 128, 155377. [Google Scholar] [CrossRef]
  28. Luo, J.; Wen, W.; Chen, J.; Zeng, X.; Wang, P.; Xu, S. Differences in tissue distribution ability of evodiamine and dehydroevodiamine are due to the dihedral angle of the molecule stereo-structure. Front. Pharmacol. 2023, 14. [Google Scholar] [CrossRef]
  29. Baltov, B.; Beyl, S.; Baburin, I.; Reinhardt, J.; Szkokan, P.; Garifulina, A.; Timin, E.; Kraushaar, U.; Potterat, O.; Hamburger, M.; et al. Assay for evaluation of proarrhythmic effects of herbal products: Case study with 12 Evodia preparations. Toxicol. Rep. 2023, 10, 589–599. [Google Scholar] [CrossRef]
  30. Hu, Q.; Chen, Y.; Zhang, W.; Li, Y.; Deng, X.; Chen, L.; Zhou, Z.; Ma, X.; Zeng, J.; Zhao, Y. Dehydroevodiamine targeting IKKβ to alleviate acute gastric injury via inhibiting the p65/NLRP3 axis. Phytomedicine 2024, 134, 155963. [Google Scholar] [CrossRef]
  31. Zheng, H.; Huang, L.; An, G.; Guo, L.; Wang, N.; Yang, W.; Zhu, Y. A Nanoreactor Based on Metal–Organic Frameworks With Triple Synergistic Therapy for Hepatocellular Carcinoma. Adv. Heal. Mater. 2024, 13, e2401743. [Google Scholar] [CrossRef]
  32. Shao, L.; Hu, T.; Fan, X.; Wu, X.; Zhou, F.; Chen, B.; Tan, S.; Xu, H.; Pan, A.; Liang, S.; et al. Intelligent Nanoplatform with Multi Therapeutic Modalities for Synergistic Cancer Therapy. ACS Appl. Mater. Interfaces 2022, 14, 13122–13135. [Google Scholar] [CrossRef] [PubMed]
  33. Han, G.; Xuewu, G.; Meng, Z.; Yuejing, W.; Yuchun, W.; Keshuang, Z.; Hongbo, Y. Therapeutic effect of dihydroartemisinin on Alzheimer’s disease model mice with senile macular degeneration. Eur. J. Med Res. 2025, 30, 1–13. [Google Scholar] [CrossRef]
  34. Bader, S.; Wilmers, J.; Pelzer, M.; Jendrossek, V.; Rudner, J. Activation of anti-oxidant Keap1/Nrf2 pathway modulates efficacy of dihydroartemisinin-based monotherapy and combinatory therapy with ionizing radiation. Free. Radic. Biol. Med. 2021, 168, 44–54. [Google Scholar] [CrossRef] [PubMed]
  35. Liang, S.-Y.; Zeng, Y.-C.; Jiang, Q.-Q.; Wu, J.-H.; Wu, Z.-Z. Pharmacokinetic studies of multi-bioactive components in rat plasma after oral administration of Xintiantai Ⅰ extract and effects of guide drug borneol on pharmacokinetics. Chin. Herb. Med. 2019, 12, 79–87. [Google Scholar] [CrossRef] [PubMed]
  36. Xiang, Z.; Guan, H.; Xie, Q.; Hu, X.; Liu, W.; Zhang, S.; Chen, Q.; Lei, J.; Shen, Q.; Liu, W.; et al. Exploring the tissue distribution propensity of active alkaloids in normal and stomach heat syndrome rats following oral administration of Zuojin Pill based on pharmacokinetics and mass spectrometry imaging. J. Ethnopharmacol. 2025, 346, 119627. [Google Scholar] [CrossRef]
  37. Wei, Y.; Ren, S.; Wang, J.; Wang, Y.; Cui, Y.; Tian, M.; Wang, R.; Liu, H.; Zhao, Y. Dehydroevodiamine ameliorates indomethacin-induced gastric injury via inhibition of ERK and p38 signaling pathway. Phytomedicine 2021, 93, 153764. [Google Scholar] [CrossRef]
  38. Jeon, H.D.; Han, Y.-H.; Mun, J.-G.; Yoon, D.H.; Lee, Y.G.; Kee, J.-Y.; Hong, S.-H. Dehydroevodiamine inhibits lung metastasis by suppressing survival and metastatic abilities of colorectal cancer cells. Phytomedicine 2022, 96, 153809. [Google Scholar] [CrossRef]
  39. Xiao, Y.; Huang, W.; Zhu, D.; Wang, Q.; Chen, B.; Liu, Z.; Wang, Y.; Liu, Q. Cancer cell membrane-camouflaged MOF nanoparticles for a potent dihydroartemisinin-based hepatocellular carcinoma therapy. RSC Adv. 2020, 10, 7194–7205. [Google Scholar] [CrossRef]
  40. Guo, W.; Liu, Y.; Chen, B.; Fan, L. Target prediction and potential application of dihydroartemisinin on hepatocarcinoma treatment. Naunyn-Schmiedeberg's Arch. Pharmacol. 2024, 397, 7711–7724. [Google Scholar] [CrossRef]
  41. Cui, Z.; Wang, H.; Li, S.; Qin, T.; Shi, H.; Ma, J.; Li, L.; Yu, G.; Jiang, T.; Li, C. Dihydroartemisinin enhances the inhibitory effect of sorafenib on HepG2 cells by inducing ferroptosis and inhibiting energy metabolism. J. Pharmacol. Sci. 2022, 148, 73–85. [Google Scholar] [CrossRef] [PubMed]
  42. Wang, Y.; Yuan, X.; Ren, M.; Wang, Z. Ferroptosis: A New Research Direction of Artemisinin and Its Derivatives in Anti-Cancer Treatment. Am. J. Chin. Med. 2024, 52, 161–181. [Google Scholar] [CrossRef] [PubMed]
  43. Jamshaid, F.; Dai, J.; Yang, L.X. New Development of Novel Berberine Derivatives against Bacteria. Mini-Reviews Med. Chem. 2020, 20, 716–724. [Google Scholar] [CrossRef] [PubMed]
  44. Rakotondrabe, T.F.; Fan, M.-X.; Muema, F.W.; Guo, M.-Q. Modulating Inflammation-Mediated Diseases via Natural Phenolic Compounds Loaded in Nanocarrier Systems. Pharmaceutics 2023, 15, 699. [Google Scholar] [CrossRef]
  45. Sultana, S.; Gupta, J.; Sharma, V.; Gupta, K.; Khosla, G.; Mishra, D. Advanced Engineered Nanoplatforms to Overcome Biological Barriers for Targeting Brain Tumors. Curr. Cancer Drug Targets 2025, 25, 1–19. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2026 MDPI (Basel, Switzerland) unless otherwise stated