Current Issue

2025 Vol. 15, No. 9

Original article
RCAN-DDI: Relation-aware cross adversarial network for drug-drug interaction prediction
Yuanyuan Zhang, Xiaoyu Xu, Baoming Feng, Haoyu Zheng, Ci'ao Zhang, Wentao Xu, Zengqian Deng
2025, 15(9) doi: 10.1016/j.jpha.2024.101159
Abstract:

Drug-drug interaction (DDI) refers to the interaction between two or more drugs in the body, altering their efficacy or pharmacokinetics. Fully considering and accurately predicting DDI has become an indispensable part of ensuring safe medication for patients. In recent years, many deep learning-based methods have been proposed to predict DDI. However, most existing computational models tend to oversimplify the fusion of drug structural and topological information, often relying on methods such as splicing or weighted summation, which fail to adequately capture the potential complementarity between structural and topological features. This loss of information may lead to models that do not fully leverage these features, thus limiting their performance in DDI prediction. To address these challenges, we propose a relation-aware cross adversarial network for predicting DDI, named RCAN-DDI, which combines a relationship-aware structure feature learning module and a topological feature learning module based on DDI networks to capture multimodal features of drugs. To explore the correlations and complementarities among different information sources, the cross-adversarial network is introduced to fully integrate features from various modalities, enhancing the predictive performance of the model. The experimental results demonstrate that the RCAN-DDI method outperforms other methods. Even in cases of labelled DDI scarcity, the method exhibits good robustness in the DDI prediction task. Furthermore, the effectiveness of the cross-adversarial module is validated through ablation experiments, demonstrating its superiority in learning multimodal complementary information.

Lipidome atlas of human myometrium reveals distinctive lipid signatures associated with adenomyosis: Combination of high-coverage lipidomics and mass spectrometry imaging
Shuo Liang, Jialin Liu, Maokun Liao, Dandan Liang, Yiyi Gong, Bo Zhang, Nan Zhao, Wei Song, Honghui Shi
2025, 15(9) doi: 10.1016/j.jpha.2025.101197
Abstract:

Adenomyosis is a common gynecological disease characterized by the invasion of endometrial glands and stroma into the myometrium of uterus, the pathological mechanism of which remains unclear yet. Disturbed lipid metabolism extensively affects abnormal cell proliferation and invasion in various diseases. However, the lipidome signature of human myometrium, which could be crucial in the development of adenomyosis, remains unknown. In this study, we generated the first lipidome profiling of human myometrium using a high-coverage and quantitative lipidomics approach based on ultra-performance liquid chromatography (UPLC) coupled with triple quadrupole (QqQ)-mass spectrometry (MS). A total of 317 lipid species were successfully quantified in the myometrial tissues from women with (n = 38) or without (n = 65) adenomyosis who underwent hysterectomy at Peking Union Medical College Hospital (Bejing, China). Up to 83 lipid species showed significant alternations in content between the two groups. These lipid aberrations involved multiple metabolic pathways, and emphasized inflammation, cell migration, and immune dysregulation upon adenomyosis. Moreover, receiver operating characteristic (ROC) curve analysis found that the combination of five lipid species could accurately distinguished the myometrial samples from women with and without adenomyosis with an area under the curve (AUC) of 0.906. Desorption electrospray ionization MS imaging (MSI) further underscored the heterogeneous distributions of these lipid markers in the adenomyosis lesion and adjacent myometrial tissue. Collectively, these results extremely improved our understanding on the molecular basis of adenomyosis, and could shed light on developing potential biomarkers and new therapeutic directions for adenomyosis.

Real-time monitoring and in vivo visualization of acetylcholinesterase activity with a near-infrared fluorescent probe
Keyun Zeng, Fang Fan, Yuqi Tang, Xiaoyu Wang, Diya Lv, Jieman Lin, Yuxin Zhang, Yingying Zhu, Yifeng Chai, Xiaofei Chen, Quan Li
2025, 15(9) doi: 10.1016/j.jpha.2025.101204
Abstract:

Acetylcholinesterase (AChE) plays a crucial role in the activities of the nervous system, and its abnormal function can lead to the occurrence and development of neurodegenerative diseases. Hence, an effective method for real-time monitoring of AChE activity is essential. Very recently, several fluorescence sensors have been developed for the detection of AChE activity, but they are usually imaging in the visible region, relatively small Stokes shifts, or long response times, limiting their application for real-time monitoring in vivo. Inspired by that, a near-infrared (NIR) off-on probe ((E)-4-(2-(4-(dicyanomethylene)-4H-chromen-2-yl)vinyl)phenyl dimethylcarbamate, DCM-N) for AChE monitoring with high selectivity and sensitivity is developed. In the probe DCM-N, a bright near-infrared fluorescence emission at 700 nm can be triggered by AChE through the cleavage of amino ester bond in DCM-N, and the resulting fluorescence exhibits a good linear relationship with AChE activity in the range of 0.2–16 U/mL, with a detection limit as low as 0.06 U/mL. For real plasma sample detection, DCM-N demonstrates advantages of accurate detection and fast response compared to the traditional Ellman assay for AChE detection. Moreover, DCM-N can be used for imaging of AChE activity in live cells and tracking of AChE activity in zebrafish models, which is of great significance for medical and physiological research related to AChE. DCM-N possesses several notable features such as light-up NIR emission, fast response, large spectral shifts and strong photostability under physiological conditions. These features enable it to monitor AChE activity both in vivo and in vitro, providing a suitable tool for real-time monitoring and in vivo visualization of AChE activity.

Mechanistic insights into honey-boiled detoxification of ChuanWu: A study on alkaloid transformation and supramolecular aggregation
Yu Zheng, Nina Wei, Chang Lu, Weidong Li, Xiaobin Jia, Linwei Chen, Rui Chen, Zhipeng Chen
2025, 15(9) doi: 10.1016/j.jpha.2025.101205
Abstract:

ChuanWu (CW), the dried mother root of Aconitum carmichaelii Debx., is a well-known traditional Chinese medicine (TCM) recognized for its potent efficacy but inherent toxicity, primarily due to its alkaloid content. Traditional and modern detoxification methods for CW include proper processing, rational compatibility, and specialized decoction techniques, among which honey-boiled CW is particularly distinctive. However, research on the detoxification mechanism of honey-boiled CW remains limited. This study investigated this mechanism by analyzing alkaloid transformation and supramolecular aggregation. Honey-boiled and water-boiled CW preparations were compared. Ultra-high-performance liquid chromatography-tandem mass spectrometry was used to analyze CW alkaloids, specifically diester alkaloids (DDAs), monoester alkaloids (MDAs), and non-esterified diterpenoid alkaloids (NDAs). Transmission electron microscopy was employed to observe and identify supramolecular aggregates in the honey-boiled CW decoction. In vivo absorption of water-boiled, honey-boiled, and NADES-boiled CW was compared. Median lethal dose (LD50) tests assessed toxicity, including hepatotoxicity and nephrotoxicity. In vitro experiments evaluated the safety, anti-inflammatory, and analgesic effects of CW-medicated serum on RAW264.7 cells, with in vivo validation in mice. Results showed that honey promoted the conversion of highly toxic DDAs to less toxic MDAs and prevented MDAs from hydrolyzing into NDAs. Honey-boiled CW formed approximately 250 nm supramolecular aggregates that encapsulated MDAs, inhibiting their conversion to NDAs. These encapsulated MDAs acted as a stable delivery system with higher bioavailability than free benzoylmesaconine. Subsequent mouse experiments confirmed that honey-boiled CW significantly increased the LD50 of CW while reducing hepatotoxicity and nephrotoxicity. Additionally, honey-boiled CW significantly improved cell safety and enhanced anti-inflammatory and analgesic effects. Our findings reveal that honey-boiled CW exhibits a potent detoxification mechanism by influencing alkaloid transformation and facilitating the formation of supramolecular aggregates. This study lays the groundwork for developing detoxification or synergistic strategies within honey-boiled TCM.

A cascade reaction nanoplatform with magnetic resonance imaging capability for combined photothermal/chemodynamic/gas cancer therapy
Jinyu Wang, Yuhao Guo, Xiaomei Wu, Yiming Ma, Qianqian Qiao, Linwei Li, Tao Liao, Ying Kuang, Cao Li
2025, 15(9) doi: 10.1016/j.jpha.2025.101223
Abstract:

To effectively exploit the tumor microenvironment (TME), TME-responsive nanocarriers based on cascade reactions have received much attention. In this study, we designed a novel nanoparticle PB@SiO2@MnO2@P-Arg (PMP) to construct a cascade reaction nanoplatform. While using biosafety Prussian blue (PB) for photothermal therapy (PTT), this nanoplatform uses silica (SiO2) as an intermediate layer to assemble Prussian blue and manganese dioxide (MnO2) into a core-shell structure, which effectively enhances the response of the nanoplatform to TME and promotes the effect of chemodynamic therapy (CDT) resulting from glutathione (GSH) depletion and Fenton-like reaction. The released Mn2+ can also be used for magnetic resonance imaging (MRI). Through the cascade reaction, poly-l-arginine (P-Arg) coated on the surface of the nanoparticles can react with hydroxyl radical (•OH) obtained from the Fenton-like reaction to release nitric oxide (NO), which further reacts with O2 to produce the more toxic peroxynitrite anion (ONOO). The photothermal effect of PB further enhances the effect of the cascade reaction while reducing the amount of heat required for treatment. In vitro and in vivo studies confirmed the antitumor effects of cascade reaction-based nanoplatforms in combined photothermal/chemodynamic/gas cancer therapies, providing new strategies for the design and fabrication of multifunctional nanoplatforms that integrate diagnostic and therapeutic functions, as well as the application of cascade reactions in multimodal synergistic therapy.

ZFP36 promotes ferroptosis and mitochondrial dysfunction and inhibits malignant progression in osteosarcoma by regulating the E2F1/ATF4 axis
Shiyue Qin, Hongyang Kong, Lei Jiang
2025, 15(9) doi: 10.1016/j.jpha.2025.101228
Abstract:

Zinc finger protein 36 (ZFP36) was found to be downregulated in osteosarcoma (OS) tumor tissues. We aimed to investigate the roles and mechanisms of ZFP36 in ferroptosis regulation during OS development. Two Gene Expression Omnibus (GEO) datasets showed that ZFP36 was a differentially expressed gene (DEG) in OS. Western blot and immunohistochemistry results showed that ZFP36 was downregulated in OS tumors and cell lines. ZFP36 overexpression plasmids and small interfering RNAs (siRNAs) were respectively transfected into OS cells. ZFP36 overexpression restrained proliferation, migration, and invasion in MG63 and U2OS cells, while ZFP36 knockdown displayed the opposite results. Moreover, ZFP36 overexpression increased the levels of intracellular Fe2+, reactive oxygen species (ROS), and malondialdehyde (MDA), and decreased the levels of glutathione (GSH), glutathione peroxidase 4 (GPX4), and solute carrier family 7 member 11 (SLC7A11). ZFP36 overexpression disturbed mitochondrial membrane potential (MMP) and mitochondrial morphology in OS cells. However, ZFP36 knockdown had the opposite results. Mechanistic studies indicated that ZFP36 promoted E2F transcription factor 1 (E2F1) messenger RNA (mRNA) degradation by binding to the AU-rich elements (AREs) within E2F1 3′ untranslated region (3′UTR) in OS cells. E2F1 overexpression abrogated the effects of ZFP36 overexpression on malignant progression, ferroptosis, and mitochondrial dysfunction in OS cells. Furthermore, E2F1 promoted the transcription activation of activating transcription factor 4 (ATF4) by binding to ATF4 promoter. E2F1 knockdown inhibited malignant progression, and promoted ferroptosis and mitochondrial dysfunction in OS cells, which was abrogated by ATF4 overexpression. Additionally, MG63 cells transfected with lentivirus ZFP36 overexpression vector (Lv-ZFP36) were injected into nude mice and tumor growth was monitored. ZFP36 overexpression significantly suppressed OS tumor growth under in vivo settings. In conclusion, ZFP36 overexpression promoted ferroptosis and mitochondrial dysfunction and inhibited malignant progression in OS by regulating the E2F1/ATF4 axis. We may provide the promising ZFP36 target for OS treatment.

Trace component fishing strategy based on offline two-dimensional liquid chromatography combined with PRDX3-surface plasmon resonance for Uncaria alkaloids
Hui Ni, Zijia Zhang, Ye Lu, Yaowen Liu, Yang Zhou, Wenyong Wu, Xinqin Kong, Liling Shen, Sihan Chen, Huali Long, Cheng Luo, Hao Zhang, Jinjun Hou, Wanying Wu
2025, 15(9) doi: 10.1016/j.jpha.2025.101244
Abstract:

The rapid screening of bioactive constituents within traditional Chinese medicine (TCM) presents a significant challenge to researchers. Prevailing strategies for the screening of active components in TCM often overlook trace components owing to their concealment by more abundant constituents. To address this limitation, a fishing strategy based on offline two-dimensional liquid chromatography (2D-LC) combined with surface plasmon resonance (SPR) was utilized to screen bioactive trace components targeting peroxiredoxin 3 (PRDX3), using Uncaria alkaloids (UAs) as a case study. Initially, an orthogonal preparative offline 2D-LC system combining a positively charged C18 column and a conventional C18 column under disparate mobile phase conditions was constructed. To fully reveal the trace alkaloids, 13 2D fractions of UAs were prepared, and their components were characterized using mass spectrometry (MS). Subsequently, employing PRDX3 as the targeting protein, a SPR-based screening approach was established and rigorously validated with geissoschizine methyl ether (GSM) serving as a positive control for binding. Employing this refined strategy, 29 candidate binding alkaloids were fished from the 13 2D fractions. Notably, combining offline 2D-LC with SPR increased the yield of candidate binding components from 10 to 29 when compared to SPR-based screening alone. Subsequent binding affinity assays confirmed that PRDX3 was a direct binding target for the 12 fished alkaloids, with isovallesiachotamine (IV), corynoxeine N-oxide (CO-N), and cadambine (CAD) demonstrating the highest affinity for PRDX3. Their interactions were further validated through molecular docking analysis. Subsequent intracellular H2O2 measurement assays and transfection experiments confirmed that these three trace alkaloids enhanced PRDX3-mediated H2O2 clearance. In conclusion, this study introduced an innovative strategy for the identification of active trace components in TCM. This approach holds promise for accelerating research on medicinal components within this field.

Naringenin: A potential therapeutic agent for modulating angiogenesis and immune response in hepatocellular carcinoma
Wenmei Wu, Xiangyu Qiu, Xiaofan Ye, Zhiliang Zhang, Siguo Xu, Xiuqi Yao, Yinyi Du, Geyan Wu, Rongxin Zhang, Jinrong Zhu
2025, 15(9) doi: 10.1016/j.jpha.2025.101254
Abstract:

Naringenin (4,5,7-trihydroxyflavonoid) is a naturally occurring bioflavonoid found in citrus fruits, which plays an important role in metabolic syndrome, neurological disorders, and cardiovascular diseases. However, the pharmacological mechanism and biological function of naringenin on anti-angiogenesis and anti-tumor immunity have not yet been elucidated. Our study firstly demonstrates that naringenin inhibits the growth of hepatocellular carcinoma (HCC) cells both in vivo and in vitro. Naringenin diminishes the ability of HCC cells to induce tube formation and migration of human umbilical vein endothelial cells (HUVECs) and suppresses neovascularization in chicken chorioallantoic membrane (CAM) assays. Meanwhile, in vivo results demonstrate that naringenin can significantly upregulate level of CD8+ T cells, subsequently increasing the level of immune-related cytokines in the tumor immune microenvironment. Mechanistically, we found that naringenin facilitate the K48-linked ubiquitination and subsequent protein degradation of vascular endothelial growth factor A (VEGFA) and mesenchymal-epithelial transition factor (c-Met), which reduces the expression of programmed death ligand 1 (PD-L1). Importantly, combination therapy naringenin with PD-L1 antibody or bevacizumab provided better therapeutic effects in liver cancer. Our study reveals that naringenin can effectively inhibit angiogenesis and anti-tumor immunity in liver cancer by degradation of VEGFA and c-Met in a K48-linked ubiquitination manner. This work enlightens the potential effect of naringenin as a promising therapeutic strategy against anti-angiogenesis and anti-tumor immunity in HCC.

Targeting Atf4 for enhanced neuroprotection: Role of quercetin-loaded EVs in ischemic stroke
Lanqing Zhao, Yu Chen, Xiaoxu Ding, Hongxi Li, Jinwei Li
2025, 15(9) doi: 10.1016/j.jpha.2025.101312
Abstract:

This study investigates the neuroprotective potential of extracellular vesicles (EVs) delivering quercetin-3-O-β-d-glucuronic acid (QG-EVs) in cerebral ischemia-reperfusion injury (CIRI). Targeted brain delivery of QG-EVs was confirmed, with neuron cells identified as pivotal in modulating CIRI through single-cell RNA sequencing (scRNA-seq). Activating transcription factor 4 (Atf4) was highlighted as a critical regulatory factor, and in vitro studies revealed that silencing Atf4 diminished the neuroprotective effects of QG-EVs, increasing oxidative stress levels and neuronal apoptosis. In a CIRI mouse model, the knockdown of Atf4 attenuated the protective outcomes provided by QG-EVs, further affirming the role of Atf4 in mediating neuroprotection. Behavioral assessments and protein analysis showed that QG-EVs significantly reduced neuronal damage and pro-apoptotic markers, while improving neurological function via Atf4 upregulation. The outcomes hint at the potential of QG-EVs as a beneficial therapeutic modality to mitigate neuronal damage in CIRI by enhancing Atf4 expression, highlighting its potential for improving ischemic stroke outcomes.

Traditional Chinese medicine-facilitated redox-labile paclitaxel dimer nanoprodrug for efficient chemoimmunotherapy
Fan Li, Wenrui Wang, Weisheng Xu, WanYing Li, Yudi Lu, Rui Wang, Zhonggui He, Zhihui Feng, Jiabing Tong, Zhenbao Li
2025, 15(9) doi: 10.1016/j.jpha.2025.101348
Abstract:

Various therapeutic modalities have been engineered for lung cancer treatment, but their clinic application is severely impeded by the poor therapy efficiency and immunosuppressive microenvironment. Herein, we fabricated a library of small molecule redox-labile nanoparticles (NPs) (i.e., diPTX-2C NPs, diPTX-2S NPs, and diPTX-2Se NPs) by the self-assembly of dimer paclitaxel (PTX) prodrug, and then utilized these NPs with the traditional Chinese medicine (TCM) Qi-Yu-San-Long-Fang (Q) for effective chemoimmunotherapy on Lewis lung carcinoma (LLC)-bearing mice models. Under the high concentration of glutathione (GSH) and H2O2, diPTX-2Se NPs could specifically release PTX in cancer cells and exert a higher selectivity and toxicity than normal cells. In LLC tumor-bearing mice, oral administration of Q not only effectively downregulated programmed death ligand-1 (PD-L1) expression, but also remodeled the immunosuppressive tumor immune microenvironment via the increase of CD4+ T and CD8+ T cell proportion and the repolarization of M2 into M1 macrophages in tumor tissues, collectively achieving superior synergistic treatment outcomes in combination with intravenous PTX prodrug NPs. Besides, we found that the combination regimen also demonstrated excellent chemoimmunotherapeutic performances on low-dose small established tumor and high-dose large established tumor models. This study may shed light on the potent utilization of Chinese and Western-integrative strategy for efficient tumor chemoimmunotherapy.

Validation of breast cancer as a risk factor for anxiety and depression: Insights from Mendelian randomization analysis
Guannan He, Man Xi, Tianhao Zhang, Shuang Wang, Gang Liu
2025, 15(9) doi: 10.1016/j.jpha.2025.101378
Abstract:

This study employed Mendelian randomization (MR) analysis to confirm the association between breast cancer and the risk of anxiety and depression, and to explore the molecular mechanisms by which lipid nanoparticles of ketamine (LNP@Ket) modulate these behaviors in a mouse model of breast cancer. Through single-cell transcriptomic analysis, the study aimed to clarify nuclear factor erythroid 2-related factor 2 (Nrf2)’s role in the development of anxiety and depression in these mice. Analysis of patient data from genome-wide association study (GWAS) databases supported the link between breast cancer, anxiety, and depression. In vivo experiments demonstrated that treating breast cancer mice with LNP@Ket significantly reduced anxiety and depression behaviors. The synthesis of LNP@Ket and its subsequent analysis highlighted its inhibitory effects on these behaviors. Single-cell transcriptomic sequencing identified key cells and genes affected by LNP@Ket treatment, particularly emphasizing Nrf2. Upregulation of Nrf2 in astrocytes increased the expression of antioxidant enzymes and reduced pro-inflammatory cytokines, alleviating anxiety and depression symptoms by inhibiting neuroinflammation and neurodegeneration. This comprehensive study highlights the pivotal role of Nrf2 in the therapeutic efficacy of LNP@Ket for treating anxiety and depression in breast cancer mice.

Review paper
Zebrafish as a vertebrate model for high-throughput drug toxicity screening: Mechanisms, novel techniques, and future perspectives
Wenhao Wang, Xuan Gao, Lin Liu, Sheng Guo, Jin-ao Duan, Ping Xiao
2025, 15(9) doi: 10.1016/j.jpha.2025.101195
Abstract:

Drug toxicity is closely related to both clinical drug safety and new drug development. Therefore, it is vital to understand the mechanisms of drug toxicity fully and to use appropriate research models with advanced technologies. Zebrafish has become an important vertebrate animal model for high-throughput drug screening and toxicity assessment. At the same time, zebrafish has an intact biological complexity, reflecting the whole organism's toxicity, which gives it an advantage over other high-throughput models in toxicity studies. Despite the gradual increase in toxicity studies utilizing zebrafish, a comprehensive and systematic review of the underlying mechanisms and new techniques is still lacking. This review aims to analyze common toxicity mechanisms in zebrafish models, such as oxidative stress, endoplasmic reticulum stress, inflammation, and apoptosis, and macroscopic changes in biological processes like lipid metabolism disorders and neurotransmitter expression abnormalities. It also introduces new technologies applied in toxicity assessment, such as gene editing, novel fluorescence imaging technology, 3D imaging technology, and novel automated technology for high-throughput screening, such as fish capsules. In addition, it also summarizes the advantages and disadvantages of the model. By doing so, it will provide new suggestions for the development and improvement of the model, make it better serve the toxicity study of clinical drugs and provide a more comprehensive perspective for drug toxicity study, thus promoting the development of the field of drug toxicity study.

Unveiling the “Dark Matter” of platelet involvement in tumor microenvironment
Peiyin Zhang, Ruiling Zu, Xingmei Zhang, Hanxiao Ren, Lubei Rao, Dongsheng Wang, Tian Li, Ping Leng, Huaichao Luo
2025, 15(9) doi: 10.1016/j.jpha.2025.101218
Abstract:

Platelets are well-known for their functions in blood clotting and vascular repair. However, in recent years, the regulatory role of platelets in the occurrence and development of malignant tumors has received significant attention. While extensive research has been conducted on the regulation of tumors by circulating platelets in peripheral blood, there is a lack of coherence and continuity among these studies. The tumor microenvironment encompasses the intricate network of cellular and acellular elements that surround and interact with tumor cells, creating a supportive ecosystem for their survival and growth. It plays a crucial role in the initiation and progression of tumors. Similar to dark matter in the universe, platelets, as tiny and enigmatic entities, play an essential role in tumor development and treatment within the tumor microenvironment. Although our current understanding of platelet regulation in the tumor microenvironment is limited, they hold immense untapped potential. In-depth studies on the tumor microenvironment have revealed platelets as a meaningful component, influencing various aspects of tumor development, metastasis, and immune evasion. Platelets, through the release of various bioactive substances or direct interaction with tumor cells, impact tumor progression while being influenced by the tumor in return. Therefore, understanding the role and mechanisms of platelets in the tumor microenvironment is of great importance for tumor prevention and treatment. This review provides a summary of the research progress on the interplay between platelets and tumors in the tumor microenvironment, and presents a promising outlook on the potential of platelets in tumor therapy.

Novel hormone therapies for advanced prostate cancer: Understanding and countering drug resistance
Zhipeng Wang, Jie Wang, Dengxiong Li, Ruicheng Wu, Jianlin Huang, Luxia Ye, Zhouting Tuo, Qingxin Yu, Fanglin Shao, Dilinaer Wusiman, William C. Cho, Siang Boon Koh, Wei Xiong, Dechao Feng
2025, 15(9) doi: 10.1016/j.jpha.2025.101232
Abstract:

Prostate cancer is the most prevalent malignant tumor among men, ranking first in incidence and second in mortality globally. Novel hormone therapies (NHT) targeting the androgen receptor (AR) pathway have become the standard of care for metastatic prostate cancer. This review offers a comprehensive overview of NHT, including abiraterone, enzalutamide, apalutamide, darolutamide, and rezvilutamide, which have demonstrated efficacy in delaying disease progression and improving patient survival and quality of life. Nevertheless, resistance to NHT remains a critical challenge. The mechanisms underlying resistance are complex, involving AR gene amplification, mutations, splice variants, increased intratumoral androgens, and AR-independent pathways such as the glucocorticoid receptor, neuroendocrine differentiation, DNA repair defects, autophagy, immune evasion, and activation of alternative signaling pathways. This review discusses these resistance mechanisms and examines strategies to counteract them, including sequential treatment with novel AR-targeted drugs, chemotherapy, poly ADP-ribose polymerase inhibitors, radionuclide therapy, bipolar androgen therapy, and approaches targeting specific resistance pathways. Future research should prioritize elucidating the molecular basis of NHT resistance, optimizing existing therapeutic strategies, and developing more effective combination regimens. Additionally, advanced sequencing technologies and resistance research models should be leveraged to identify novel therapeutic targets and improve drug delivery efficiencies. These advancements hold the potential to overcome NHT resistance and significantly enhance the management and prognosis of patients with advanced prostate cancer.

Research progress and challenges of molecular recognition techniques in the screening of active ingredients in traditional Chinese medicine
Lin Li, Qi Li, Yanxiao Li, Dandan Gong, Bonian Zhao
2025, 15(9) doi: 10.1016/j.jpha.2025.101243
Abstract:

Traditional Chinese medicine (TCM) has become an important treasure trove of natural resources for the development of new medicines due to their diverse compositions, significant therapeutic effects, and few side effects. The screening of active ingredients in TCM represents a crucial step in elucidating the material basis and mechanism of action of TCM. At present, efficient and precise molecular recognition techniques based on intermolecular interactions have been extensively employed for the identification of active ingredients in TCM. This paper presents a review of the fundamental principles underlying solution-phase/affinity ligand fishing, solid-phase/affinity ligand fishing, molecular imprinting and molecular docking techniques, with a particular focus on their applications in the screening of active ingredients in TCM. Furthermore, the paper compares the advantages and disadvantages of the various techniques and identifies the limitations of existing techniques. In conclusion, the paper identifies the prospective trajectory of molecular recognition techniques in the domain of TCM research. This paper not only provides theoretical references for the development of new methods of active ingredient screening but also helps to promote the modernization and internationalization of TCM.

Understanding the mechanistic and therapeutic perspectives on cytokines and chemokines in acute high-altitude illness syndromes
Amin Ullah, Rajeev K. Singla, Yingbo Zhang, ShanShan Hu, Bairong Shen
2025, 15(9) doi: 10.1016/j.jpha.2025.101249
Abstract:

Acute high-altitude (HA) illnesses (AHAIs), including acute mountain sickness (AMS), HA cerebral edema (HACE), and HA pulmonary edema (HAPE), represent significant health challenges for individuals rapidly ascending to high altitudes. Cytokines (interleukins (ILs)) and chemokines, which are involved in inflammatory and immunological responses, regulate the response of the body to hypoxic stress. Their dysregulation can contribute to the clinical symptoms of AMS, HACE, and HAPE by increasing vascular permeability, causing edema and damaging tissue. AHAIs elevate the levels of pro-inflammatory cytokines and chemokines, such as IL-17, tumor necrosis factor α (TNF-α), IL-1, IL-6, C−X−C motif chemokine ligand (CXCL) 10, CXCL8, C−C motif ligand 2 (CCL2), and CCL3, exacerbating symptoms. Thus, this review focuses on the cytokines and chemokines involved in AHAIs and the molecular mechanisms that extend beyond these cytokines and chemokines in clinical and preclinical contexts. Identifying these mediators and pathways helps researchers design drugs that reduce symptoms, slow disease progression, and enhance outcomes. Cytokines and chemokines have complex functions in these disorders and may serve as prospective therapeutic targets. Finally, we discuss treatment possibilities for AHAIs (drugs, exercise, and other inhibitors). This knowledge will help us to protect and improve the health of individuals at high altitudes.

Pathogenesis and treatment strategies for infectious keratitis: Exploring antibiotics, antimicrobial peptides, nanotechnology, and emerging therapies
Man Yu, Ling Li, Yijun Liu, Ting Wang, Huan Li, Chen Shi, Xiaoxin Guo, Weijia Wu, Chengzi Gan, Mingze Li, Jiaxu Hong, Kai Dong, Bo Gong
2025, 15(9) doi: 10.1016/j.jpha.2025.101250
Abstract:

Infectious keratitis (IK) is a leading cause of blindness worldwide, primarily resulting from improper contact lens use, trauma, and a compromised immune response. The pathogenic microorganisms responsible for IK include bacteria, fungi, viruses, and Acanthamoeba. This review examines standard therapeutic agents for treating IK, including broad-spectrum empiric antibiotics for bacterial keratitis (BK), antifungals such as voriconazole and natamycin for fungal infections, and antiviral nucleoside analogues for viral keratitis (VK). Additionally, this review discusses therapeutic agents, such as polyhexamethylene biguanide (PHMB), for the treatment of Acanthamoeba keratitis (AK). The review also addresses emerging drugs and the challenges associated with their clinical application, including anti-biofilm agents that combat drug resistance and nuclear factor kappa-B (NF-κB) pathway-targeted therapies to mitigate inflammation. Furthermore, methods of Photodynamic Antimicrobial Therapy (PDAT) are explored. This review underscores the importance of integrating novel and traditional therapies to tackle drug resistance and enhance drug delivery, with the goal of advancing treatment strategies for IK.

Short communication
VLDL: The key factors influencing the distribution of mitotane in patients with adrenocortical carcinoma
Xiangjie Di, Yixian Liu, Jia You, Yuchun Men, Zhenlei Wang, Chao Zhou, Ying Jin, Yating Ge, Yongji He, Li Zheng
2025, 15(9) doi: 10.1016/j.jpha.2025.101252
Abstract:
Integrating explainable deep learning with multi-omics for screening progressive diagnostic biomarkers of hepatocellular carcinoma covering the “inflammation-cancer” transformation
Saiyu Li, Yiwen Zhang, Lifang Guan, Yijing Dong, Mingzhe Zhang, Qian Zhang, Huarong Xu, Wei Xiao, Zhenzhong Wang, Yan Cui, Qing Li
2025, 15(9) doi: 10.1016/j.jpha.2025.101253
Abstract:
Commentary
A promising leap in early screening for chronic kidney disease: The smartphone-integrated handheld automated biochemical analyzer
Ming Chen
2025, 15(9) doi: 10.1016/j.jpha.2025.101436
Abstract:
Perspective
Toward realistic pharmaceutical evaluation: Challenges in 3D-printed organ chip of vascularized tissue with microcirculation
Ling Wang, Ziyan Qiu, Yingjie Liu, Fei Wang, Dichen Li
2025, 15(9) doi: 10.1016/j.jpha.2025.101445
Abstract: