1
|
Albassam H, Almutairi O, Alnasser M, Altowairqi F, Almutairi F, Alobid S. Discovery of a selective PI3Kα inhibitor via structure-based virtual screening for targeted colorectal cancer therapy. J Enzyme Inhib Med Chem 2025; 40:2468852. [PMID: 39992303 PMCID: PMC11852364 DOI: 10.1080/14756366.2025.2468852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/31/2025] [Accepted: 02/08/2025] [Indexed: 02/25/2025] Open
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related mortality globally, driving an urgent need for effective therapies. A promising avenue of research focuses on the PI3K/AKT/mTOR signalling pathway, which is frequently disrupted by mutations in the PI3Kα subunit. Our cutting-edge study employed a structure-based virtual screening of ∼3000 compounds, leading to the discovery of F0608-0019, a highly potent and selective PI3Kα inhibitor. F0608-0019 demonstrated remarkable efficacy in suppressing HCT116 colorectal cancer cell proliferation, with an IC50 of 12.14 µM, while maintaining high selectivity by minimising activity against other PI3K isoforms. Advanced molecular dynamics simulations highlighted the stability of F0608-0019's binding interactions with key amino acids, such as TRP:780, ILE:932, and VAL:850, which are critical for its targeted action. These exciting findings reveal F0608-0019 as a leading candidate for innovative CRC therapies that selectively target PI3Kα dysregulation, offering promising new possibilities for effective CRC treatment.
Collapse
Affiliation(s)
- Hussam Albassam
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Omar Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Majed Alnasser
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Faisal Altowairqi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Faris Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Saad Alobid
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
2
|
Chavan PR, Pandey R, Patil BM, Murti K, Kumar N. Unravelling key signaling pathways for the therapeutic targeting of non-small cell lung cancer. Eur J Pharmacol 2025; 998:177494. [PMID: 40090536 DOI: 10.1016/j.ejphar.2025.177494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/18/2025]
Abstract
Lung cancer (LC) remains the foremost cause of cancer-related mortality across the globe. Non-small cell lung cancer (NSCLC) is a type of LC that exhibits significant heterogeneity at histological and molecular levels. Genetic alterations in upstream signaling molecules activate cascades affecting apoptosis, proliferation, and differentiation. Disruption of these signaling pathways leads to the proliferation of cancer-promoting cells, progression of cancer, and resistance to its treatment. Recent insights into the function of signaling pathways and their fundamental mechanisms in the onset of various diseases could pave the way for new therapeutic approaches. Recently, numerous drug molecules have been created that target these cell signaling pathways and could be used alongside other standard therapies to achieve synergistic effects in mitigating the pathophysiology of NSCLC. Additionally, many researchers have identified several predictive biomarkers, and alterations in transcription factors and related pathways are employed to create new therapeutic strategies for NSCLC. Findings suggest using specific inhibitors to target cellular signaling pathways in tumor progression to treat NSCLC. This review investigates the role of signaling pathways in NSCLC development and explores novel therapeutic strategies to enhance clinical treatment options for NSCLC.
Collapse
Affiliation(s)
- Pavan Ramrao Chavan
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research, Hajipur, Bihar, India
| | - Ruchi Pandey
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research, Hajipur, Bihar, India
| | - Baswant Malesh Patil
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education & Research, Hajipur, Bihar, India
| | - Krishna Murti
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education & Research, Hajipur, Bihar, India
| | - Nitesh Kumar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research, Hajipur, Bihar, India.
| |
Collapse
|
3
|
Hossain MT, Hossain MA. Targeting PI3K in cancer treatment: A comprehensive review with insights from clinical outcomes. Eur J Pharmacol 2025; 996:177432. [PMID: 40020984 DOI: 10.1016/j.ejphar.2025.177432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
The phosphoinositide 3-kinase (PI3K) pathway plays a crucial role in cancer, including cell growth, survival, metabolism, and metastasis. Its major role in tumor growth makes it a key target for cancer therapeutics, offering significant potential to slow tumor progression and enhance patient outcomes. Gain-of-function mutations, gene amplifications, and the loss of regulatory proteins like PTEN are frequently observed in malignancies, contributing to tumor development and resistance to conventional treatments such as chemotherapy and hormone therapy. As a result, PI3K inhibitors have received a lot of interest in cancer research. Several kinds of small-molecule PI3K inhibitors have been developed, including pan-PI3K inhibitors, isoform-specific inhibitors, and dual PI3K/mTOR inhibitors, each targeting a distinct component of the pathway. Some PI3K inhibitors such as idelalisib, copanlisib, duvelisib, alpelisib, and umbralisib have received FDA-approval, and are effective in the treatment of breast cancer and hematologic malignancies. Despite promising results in preclinical and clinical trials, the overall clinical success of PI3K inhibitors has been mixed. While some patients may get substantial advantages, a considerable number of them acquire resistance as a result of feedback activation of alternative pathways, adaptive tumor responses, and treatment-emergent mutations. The resistance mechanisms provide barriers to the sustained efficacy of PI3K-targeted treatments. This study reviews recent advancements in PI3K inhibitors, covering their clinical status, mechanism of action, resistance mechanisms, and strategies to overcome resistance.
Collapse
Affiliation(s)
- Md Takdir Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh.
| | - Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh.
| |
Collapse
|
4
|
Dong D, Yu X, Liu H, Xu J, Guo J, Guo W, Li X, Wang F, Zhang D, Liu K, Sun Y. Study of immunosenescence in the occurrence and immunotherapy of gastrointestinal malignancies. Semin Cancer Biol 2025; 111:16-35. [PMID: 39929408 DOI: 10.1016/j.semcancer.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/18/2025] [Accepted: 01/26/2025] [Indexed: 02/25/2025]
Abstract
In human beings heterogenous, pervasive and lethal malignancies of different parts of the gastrointestinal (GI) tract viz., tumours of the oesophagus, stomach, small intestine, colon, and rectum, represent gastrointestinal malignancies. Primary treatment modality for gastric cancer includes chemotherapy, surgical interventions, radiotherapy, monoclonal antibodies and inhibitors of angiogenesis. However, there is a need to improve upon the existing treatment modality due to associated adverse events and the development of resistance towards treatment. Additionally, age has been found to contribute to increasing the incidence of tumours due to immunosenescence-associated immunosuppression. Immunosenescence is the natural process of ageing, wherein immune cells as well as organs begin to deteriorate resulting in a dysfunctional or malfunctioning immune system. Accretion of senescent cells in immunosenescence results in the creation of a persistent inflammatory environment or inflammaging, marked with elevated expression of pro-inflammatory and immunosuppressive cytokines and chemokines. Perturbation in the T-cell pools and persistent stimulation by the antigens facilitate premature senility of the immune cells, and senile immune cells exacerbate inflammaging conditions and the inefficiency of the immune system to identify the tumour antigen. Collectively, these conditions contribute positively towards tumour generation, growth and eventually proliferation. Thus, activating the immune cells to distinguish the tumour cells from normal cells and invade them seems to be a logical strategy for the treatment of cancer. Consequently, various approaches to immunotherapy, viz., programmed death ligand-1 (PD-1) inhibitors, Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors etc are being extensively evaluated for their efficiency in gastric cancer. In fact, PD-1 inhibitors have been sanctioned as late late-line therapy modality for gastric cancer. The present review will focus on deciphering the link between the immune system and gastric cancer, and the alterations in the immune system that incur during the development of gastrointestinal malignancies. Also, the mechanism of evasion by tumour cells and immune checkpoints involved along with different approaches of immunotherapy being evaluated in different clinical trials will be discussed.
Collapse
Affiliation(s)
- Daosong Dong
- Department of Pain, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Xue Yu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Molecular Pathology and Epidemiology of Gastric Cancer in the Universities of Liaoning Province, Shenyang, Liaoning 110001, China
| | - Haoran Liu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Jingjing Xu
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Jiayan Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Wei Guo
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiang Li
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Fei Wang
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Dongyong Zhang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Kaiwei Liu
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Yanbin Sun
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
5
|
Liao JN, Ni WJ, Wu PH, Yang YD, Yang Y, Long W, Xie MZ, Zhu XZ, Xie FH, Leng XM. Switching from messenger RNAs to noncoding RNAs, METTL3 is a novel colorectal cancer diagnosis and treatment target. World J Gastrointest Oncol 2025; 17:104076. [DOI: 10.4251/wjgo.v17.i5.104076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/10/2025] [Accepted: 04/03/2025] [Indexed: 05/15/2025] Open
Abstract
N6-methyladenosine (m6A) modification, one of the most prevalent RNA epigenetic modifications in eukaryotes, constitutes over 60% of all RNA methylation modifications. This dynamic modification regulates RNA processing, maturation, nucleocytoplasmic transport, translation efficiency, phase separation, and stability, thereby linking its dysregulation to diverse physiological and pathological processes. METTL3, a core catalytic component of the methyltransferase complex responsible for m6A deposition, is frequently dysregulated in diseases, including colorectal cancer (CRC). Although METTL3’s involvement in CRC pathogenesis has been documented, its precise molecular mechanisms and functional roles remain incompletely understood. METTL3 mediates CRC progression-encompassing proliferation, invasion, drug resistance, and metabolic reprogramming-through m6A-dependent modulation of both coding RNAs and noncoding RNAs. Its regulatory effects are primarily attributed to interactions with key signaling pathways at multiple stages of CRC development. Emerging evidence highlights METTL3 as a promising biomarker for CRC diagnosis and prognosis, as well as a potential therapeutic target. By synthesizing recent advances in METTL3 research within CRC, this review provides critical insights into novel strategies for clinical diagnosis and targeted therapy.
Collapse
Affiliation(s)
- Jun-Nan Liao
- The First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| | - Wen-Juan Ni
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| | - Ping-Hui Wu
- The First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| | - Ya-Dong Yang
- The First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| | - Ying Yang
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| | - Wen Long
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| | - Mei-Zhen Xie
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| | - Xiu-Zhi Zhu
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| | - Fu-Hua Xie
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| | - Xiao-Min Leng
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| |
Collapse
|
6
|
Liu L, Wuyun T, Sun X, Zhang Y, Cha G, Zhao L. Therapeutic efficacy of TMTP1-modified EVs in overcoming bone metastasis and immune resistance in PIK3CA mutant NSCLC. Cell Death Dis 2025; 16:367. [PMID: 40328748 PMCID: PMC12055990 DOI: 10.1038/s41419-025-07685-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 04/08/2025] [Accepted: 04/17/2025] [Indexed: 05/08/2025]
Abstract
Non-small cell lung cancer (NSCLC) with PIK3CA mutations demonstrates significant challenges in treatment due to enhanced bone metastasis and immune checkpoint resistance. This study investigates the efficacy of tumor-targeting peptide 1-modified cancer stem cell-derived extracellular vesicles (TMTP1-TSRP-EVs) in reshaping the tumor microenvironment and reversing immune checkpoint resistance in NSCLC. By integrating TMTP1-TSRP into EVs, we aim to specifically deliver therapeutic agents to NSCLC cells, focusing on inhibiting the PI3K/Akt/mTOR pathway, a crucial driver of oncogenic activity and immune evasion in PIK3CA-mutated cells. Our comprehensive in vitro and in vivo analyses show that TMTP1-TSRP-EVs significantly inhibit tumor growth, reduce PD-L1 expression, and enhance CD8+ T cell infiltration, effectively reversing the immune-suppressive microenvironment. Moreover, the in vivo models confirm that our approach not only suppresses bone metastases but also overcomes primary resistance to immune checkpoint inhibitors by modulating the expression of key immunological markers. These findings suggest that targeted delivery of TMTP1-TSRP-EVs could provide a novel therapeutic strategy for treating PIK3CA-mutant NSCLC, offering significant improvements over traditional therapies by directly targeting the molecular pathogenesis of tumor resistance and metastasis. Molecular Mechanisms Reshaping the TME to Halt PI3K-Mutant Bone Metastasis of NSCLC and Overcoming Primary ICI Resistance. (Created by BioRender).
Collapse
Affiliation(s)
- Liwen Liu
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Tanghesi Wuyun
- The Second Department of Respiratory, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xin Sun
- The Second Department of Respiratory, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yu Zhang
- The Second Department of Respiratory, Harbin Medical University Cancer Hospital, Harbin, China
| | - Geqi Cha
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ling Zhao
- The Second Department of Respiratory, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
7
|
Cartwright D, Kidd AC, Ansel S, Ascierto ML, Spiliopoulou P. Oncogenic Signalling Pathways in Cancer Immunotherapy: Leader or Follower in This Delicate Dance? Int J Mol Sci 2025; 26:4393. [PMID: 40362630 DOI: 10.3390/ijms26094393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 05/01/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
Immune checkpoint inhibitors have become a mainstay of treatment in many solid organ malignancies. Alongside this has been the rapid development in the identification and targeting of oncogenic drivers. The presence of alterations in oncogenic drivers not only predicts response to target therapy but can modulate the immune microenvironment and influence response to immunotherapy. Combining immune checkpoint inhibitors with targeted agents is an attractive therapeutic option but overlapping toxicity profiles may limit the clinical use of some combinations. In addition, there is growing evidence of shared resistance mechanisms that alter the response to immunotherapy when it is used after targeted therapy. Understanding this complex interaction between oncogenic drivers, targeted therapy and response to immune checkpoint inhibitors is vital for selecting the right treatment, at the right time for the right patient. In this review, we summarise the preclinical and clinical evidence of the influence of four common oncogenic alterations on immune checkpoint inhibitor response, combination therapies, and the presence of shared resistance mechanisms. We highlight the common resistance mechanisms and the need for more randomised trials investigating both combination and sequential therapy.
Collapse
Affiliation(s)
- Douglas Cartwright
- School of Cancer Sciences, University of Glasgow, Bearsden, Glasgow G61 1QH, UK
- Beatson West of Scotland Cancer Centre,1053 Great Western Road, Glasgow G12 0YN, UK
| | - Andrew C Kidd
- School of Cancer Sciences, University of Glasgow, Bearsden, Glasgow G61 1QH, UK
- Beatson West of Scotland Cancer Centre,1053 Great Western Road, Glasgow G12 0YN, UK
| | - Sonam Ansel
- School of Cancer Sciences, University of Glasgow, Bearsden, Glasgow G61 1QH, UK
- Beatson West of Scotland Cancer Centre,1053 Great Western Road, Glasgow G12 0YN, UK
| | | | - Pavlina Spiliopoulou
- School of Cancer Sciences, University of Glasgow, Bearsden, Glasgow G61 1QH, UK
- Beatson West of Scotland Cancer Centre,1053 Great Western Road, Glasgow G12 0YN, UK
| |
Collapse
|
8
|
Sun MX, Zhu HC, Yu Y, Yao Y, Li HY, Feng FB, Wang QY, Liu RJ, Sun CG. Role of the Wnt signaling pathway in the complex microenvironment of breast cancer and prospects for therapeutic potential (Review). Int J Oncol 2025; 66:36. [PMID: 40145557 PMCID: PMC12068849 DOI: 10.3892/ijo.2025.5742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
The focus on breast cancer treatment has shifted from the cytotoxic effects of single drugs on tumor cells to multidimensional multi‑pathway synergistic intervention strategies targeting the tumor microenvironment (TME). The activation of the Wnt signaling pathway in the TME of breast cancer cells serves a key regulatory role in tissue homeostasis and is a key driver of the carcinogenic process. Modulating the crosstalk between the Wnt pathway and TME of breast cancer is key for understanding the biological behavior of breast cancer and advancing the development of novel antitumor drugs. The present review aimed to summarize the complex mechanisms of the Wnt signaling pathway in the breast cancer TME, interactions between the Wnt signaling pathway and components of the breast cancer TME and breast cancer‑associated genes, as well as the interactions between the Wnt signaling pathway and other signaling cascades at the molecular level. Furthermore, the present review aimed to highlight the unique advantages of the Wnt signaling pathway in the macro‑regulation of the TME and the current therapeutic strategies targeting the Wnt signaling pathway, their potential clinical value and future research directions in breast cancer treatment.
Collapse
Affiliation(s)
- Meng Xuan Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Han Ci Zhu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Yang Yu
- State Key Laboratory of Quality Research in Chinese Medicine, and Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, P.R. China
| | - Yan Yao
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Hua Yao Li
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Fu Bin Feng
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Qing Yang Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Rui Juan Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
| | - Chang Gang Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 261000, P.R. China
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
9
|
Yu J, Hu Q, Fan K, Gao Y, Li Y. C15orf39, a downstream effector of PI3K/AKT signaling, promotes gastric carcinogenesis and correlates with patient outcomes. Int J Biol Macromol 2025; 306:141615. [PMID: 40032127 DOI: 10.1016/j.ijbiomac.2025.141615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 02/01/2025] [Accepted: 02/27/2025] [Indexed: 03/05/2025]
Abstract
The phosphatidylinositol-3-kinases (PI3K) signaling pathway is highly complex and well-known to exert oncogenic roles in multiple cancer types. Exploring new factors involved in this pathway may offer the potential for improving the early diagnosis and treatment strategies for cancers. Here we used gastric cancer (GC) as a model to identify co-regulated effectors downstream of three catalytic subunits of PI3K through high-throughput sequencing in PIK3CA, PIK3CB, and PIK3CD knockdown GC cells. C15orf39, a new uncharacterized gene, was selected due to the most significant expression change. qRT-PCR and immunohistochemistry analyses revealed that C15orf39 was frequently upregulated in GC tissues and strongly correlated with poor clinical outcomes in GC patients. Gain- and loss-of-function studies demonstrated that C15orf39 promoted GC cell proliferation, migration, and drug resistance. Mechanistically, C15orf39 promoted GC progression possibly via modulating cell mitosis and cell cycle. FOXK2, a transcription factor activated by PI3K/AKT signaling, could bind to the promoter of C15orf39 and positively regulate C15orf39 expression. These findings unveiled a new PI3K/AKT/FOXK2/C15orf39 signaling axis that promotes GC development and progression. C15orf39 may become a potential biomarker for early diagnosis and personalized treatment to improve the prognosis of GC patients.
Collapse
Affiliation(s)
- Jiahua Yu
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Qingqing Hu
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Kailing Fan
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Yong Gao
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China.
| | - Yandong Li
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China; Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China.
| |
Collapse
|
10
|
Yarlagadda S, de Oliveira Andrade M, Nanda R. CDK4/6 inhibition in early and advanced hormone receptor-positive, HER2-negative breast cancer. Expert Rev Anticancer Ther 2025:1-12. [PMID: 40285528 DOI: 10.1080/14737140.2025.2498994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
INTRODUCTION Cyclin-dependent kinase 4 and 6 (CDK4/6) inhibition in combination with endocrine therapy is the mainstay of treatment for hormone receptor-positive, HER2-negative (HR + /HER2-) advanced breast cancer; the CDK4/6 inhibitors abemaciclib and ribociclib are also approved for high-risk, early-stage, HR + /HER2- breast cancer. Numerous studies exploring CDK4/6 inhibitors in the early-stage setting are ongoing, as well as many more exploring novel combinations in the metastatic setting. AREAS COVERED Here, we review the basis of CDK4/6 inhibition for HR +/HER2- breast cancer, the pivotal clinical trials which led to regulatory approval, and ongoing trials evaluating novel combinations to further improve outcomes for those with both early and advanced HR+/HER2- breast cancer. Current literature was reviewed by a comprehensive search of PubMed MEDLINE (1/1/2000-12/31/2024). EXPERT OPINION CDK4/6 inhibitors are integral in the management of both advanced and high-risk, early-stage HR + /HER2- breast cancer. Biomarkers predictive of CDK 4/6 inhibitor (CDK4/6i) benefit remain elusive, and clinical and pathological features remain key to identifying those who are candidates for CDK4/6 inhibition in the early-stage setting. Numerous trials evaluating the role of a CDK4/6i with novel endocrine therapy partners and other targeted agents are ongoing, with the goal of improving outcomes for those with HR + /HER2- disease.
Collapse
Affiliation(s)
- Sudha Yarlagadda
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL, USA
| | | | - Rita Nanda
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL, USA
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago Medicine Comprehensive Cancer Center, Chicago, IL, USA
| |
Collapse
|
11
|
Dawalibi A, Bakir M, Mohammad KS. The genetic architecture of bone metastases: unveiling the role of epigenetic and genetic modifications in drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:19. [PMID: 40342734 PMCID: PMC12059479 DOI: 10.20517/cdr.2025.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/26/2025] [Accepted: 04/17/2025] [Indexed: 05/11/2025]
Abstract
Bone metastases represent frequent and severe complications in various cancers, notably impacting prognosis and quality of life. This review article delves into the genetic and epigenetic mechanisms underpinning drug resistance in bone metastases, a key challenge in effective cancer treatment. The development of drug resistance in cancer can manifest as either intrinsic or acquired, with genetic heterogeneity playing a pivotal role. Intrinsic resistance is often due to pre-existing mutations, while acquired resistance evolves through genetic and epigenetic alterations during treatment. These alterations include mutations in driver genes like TP53 and RB1, epigenetic modifications such as DNA methylation and histone changes, and pathway alterations, notably involving RANK-RANKL signaling and the PI3K/AKT/mTOR cascade. Recent studies underline the significance of the tumor microenvironment in fostering drug resistance, with components such as cancer-associated fibroblasts and hypoxia playing crucial roles. The interactions between metastatic cancer cells and the bone microenvironment facilitate survival and the proliferation of drug-resistant clones. This review highlights the necessity of understanding these complex interactions to develop targeted therapies that can overcome resistance and improve treatment outcomes. Current therapeutic strategies and future directions are discussed, emphasizing the integration of genomic profiling and targeted interventions in managing bone metastases. The evolving landscape of genetic research, including the application of next-generation sequencing and CRISPR technology, offers promising avenues for novel and more effective therapeutic strategies. This comprehensive exploration aims to provide insights into the molecular intricacies of drug resistance in bone metastases, paving the way for improved clinical management and patient care.
Collapse
Affiliation(s)
- Ahmad Dawalibi
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Mohamad Bakir
- Department of Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Khalid S. Mohammad
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
12
|
Carr A, Coulter JA, Workman J, Fay J, Farrelly A, Eustace AJ, Bennie L, Grogan L, Breathnach O, Morris PG, McNamara DA, Cremona M, O'Neill BDP, Hennessy BT, Toomey S. Targeting the phosphatidylinositol-3-kinase (PI3K) and mitogen activated protein kinase (MAPK) signalling pathways to enhance chemoradiotherapy in locally advanced rectal cancer. Cancer Treat Res Commun 2025; 43:100926. [PMID: 40245445 DOI: 10.1016/j.ctarc.2025.100926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 03/04/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
Responses to neoadjuvant chemoradiotherapy for locally advanced rectal cancer are not uniform. The phosphatidylinositol-3 kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways are involved in tumorigenesis and treatment resistance in many cancers; therefore, targeting these pathways could enhance response to chemoradiotherapy. A panel of colorectal cancer (CRC) cell lines (n = 10) with varying PI3K and MAPK mutational backgrounds were treated with combinations of 5-Flourouracil (5-FU), radiation, the PI3K inhibitor copanlisib, and/or the MEK inhibitor refametinib, and their effects on proliferation in vitro were measured. BALB/c SCID mice were implanted with CRC cell lines representative of each mutational background, treated with copanlisib and/or chemoradiotherapy, and monitored for tumor growth. In vitro, PIK3CA mutated cell lines were most sensitive to copanlisib (IC50=28 nM) and KRAS mutated cell lines were most sensitive to refametinib (IC50 = 36 nM), while the combination of copanlisib and refametinib was synergistic in 9/10 cell lines tested. The addition of copanlisib to 5-FU chemoradiotherapy inhibited cell growth compared to 5-FU chemoradiotherapy alone, an effect that was most notable in LS-1034 (KRAS mutated) and Caco-2 (PIK3CA/KRAS wild-type) cell lines. In vivo copanlisib and 5-FU chemoradiotherapy reduced tumor growth in all xenograft models and increased overall survival in LS-1034 and Caco-2 xenografts. Our results suggest that activation of the kinase signalling pathway may modulate PI3K/MEK inhibitor responsiveness in colorectal cancer. Furthermore, the addition of copanlisib to 5-FU chemoradiotherapy resulted in an enhanced anti-proliferative cytotoxic effect compared to 5-FU chemoradiotherapy alone, regardless of the background mutational status, and supports further clinical development of this regimen.
Collapse
Affiliation(s)
- Aoife Carr
- Department of Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | | | - Julie Workman
- Department of Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Joanna Fay
- Department of Pathology, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Angela Farrelly
- Department of Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Alex J Eustace
- Department of Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Lindsey Bennie
- School of Pharmacy, Queen's University Belfast, Belfast, UK
| | - Liam Grogan
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland; Beaumont RCSI Cancer Centre, Dublin, Ireland
| | - Oscar Breathnach
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland; Beaumont RCSI Cancer Centre, Dublin, Ireland
| | - Patrick G Morris
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland; Beaumont RCSI Cancer Centre, Dublin, Ireland
| | - Deborah A McNamara
- Beaumont RCSI Cancer Centre, Dublin, Ireland; Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Mattia Cremona
- Department of Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Brian D P O'Neill
- Beaumont RCSI Cancer Centre, Dublin, Ireland; St. Luke's Radiation Oncology Network, Dublin, Ireland
| | - Bryan T Hennessy
- Department of Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland; Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland; Beaumont RCSI Cancer Centre, Dublin, Ireland
| | - Sinead Toomey
- Department of Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland; Beaumont RCSI Cancer Centre, Dublin, Ireland.
| |
Collapse
|
13
|
Heo Y, Kim WJ, Cho YJ, Jung JW, Kim NS, Choi IY. Advances in cancer genomics and precision oncology. Genes Genomics 2025; 47:399-416. [PMID: 39849190 DOI: 10.1007/s13258-024-01614-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 12/27/2024] [Indexed: 01/25/2025]
Abstract
BACKGROUND Next-generation sequencing has revolutionized genome science over the last two decades. Indeed, the wealth of sequence information on our genome has deepened our understanding on cancer. Cancer is a genetic disease caused by genetic or epigenetic alternations that affect the expression of genes that control cell functions, particularly cell growth and division. Utilization of next-generation sequencing in cancer gene panels has enabled the identification of actionable gene alterations in cancer patients to guide personalized precision medicine. OBJECTIVE The aim is to provide information that can identify actionable gene alterations, enabling personalized precision medicine for cancer patients. RESULTS & DISCUSSION Equipped with next-generation sequencing techniques, international collaboration programs on cancer genomics have identified numerous mutations, gene fusions, microsatellite variations, copy number variations, and epigenetics changes that promote the transformation of normal cells into tumors. Cancer classification has traditionally been based on cell type or tissue-of-origin and the morphological characteristics of the cancer. However, interactive genomic analyses have currently reclassified cancers based on systemic molecular-based taxonomy. Although all cancer-causing genes and mechanisms have yet to be completely understood or identified, personalized or precision medicine is now currently possible for some forms of cancer. Unlike the "one-size-fits-all" approach of traditional medicine, precision medicine allows for customized or personalized treatment based on genomic information. CONCLUSION Despite the availability of numerous cancer gene panels, technological innovation in genomics and expansion of knowledge on the cancer genome will allow precision oncology to manage even more types of cancers.
Collapse
Affiliation(s)
- Yonjong Heo
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, 24341, Gangwon, Republic of Korea
| | - Woo-Jin Kim
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, 24341, Gangwon, Republic of Korea
| | - Yong-Joon Cho
- Department of Molecular Bioscience, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Multidimensional Genomics Research Center, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jae-Won Jung
- Genetic Sciences Group, Thermo Fisher Scientific Solutions Korea Co., Ltd., Seoul, 06349, Republic of Korea
| | - Nam-Soo Kim
- Department of Molecular Bioscience, Kangwon National University, Chuncheon, 24341, Republic of Korea.
- NBIT Co., Ltd., Chuncheon, 24341, Republic of Korea.
| | - Ik-Young Choi
- Department of Smart Farm and Agricultural Industry, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
14
|
Dexheimer TS, Davoudi Z, Coussens NP, Silvers T, Morris J, Takebe N, Said R, Moscow JA, Doroshow JH, Teicher BA. Combinatorial screen of targeted agents with the PI3K inhibitors inavolisib, alpelisib, duvelisib, and copanlisib in multi-cell type tumor spheroids. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2025; 32:100222. [PMID: 39999911 PMCID: PMC12034487 DOI: 10.1016/j.slasd.2025.100222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/05/2025] [Accepted: 02/22/2025] [Indexed: 02/27/2025]
Abstract
Dysregulation of the phosphatidylinositol 3-kinase (PI3K) pathway is a key contributor to cancer, making PI3K inhibitors a promising approach for targeted therapy. The selectivity of available inhibitors varies across different PI3K isoforms. Alpelisib and inavolisib are selective for the α-isoform, while duvelisib targets the δ- and γ-isoforms, and copanlisib is a pan-PI3K inhibitor, active against all isoforms. This study investigated the activity of these four PI3K inhibitors in combination with other targeted agents using multi-cell type tumor spheroids composed of 60% malignant cells, 25% endothelial cells, and 15% mesenchymal stem cells. Twenty-nine tumor spheroid models were evaluated, including twenty-six patient-derived cancer cell lines from the NCI Patient-Derived Models Repository and three established cell lines from the NCI-60 human tumor cell line panel. Additive and/or synergistic effects were observed with alpelisib or inavolisib or copanlisib in combination with a RAS/MEK/ERK pathway inhibitor, either selumetinib (MEK), ravoxertinib (ERK 1/2), or tovorafenib (DAY101, RAF). Combinations of each of these three PI3K inhibitors with the KRAS mutation specific inhibitors MTRX1133 (KRAS G12D) or sotorasib (KRAS G12C) had selective activity in cell lines harboring the corresponding target. Lastly, combination effects were observed from vertical inhibition of the PI3K/AKT/mTOR pathway with a PI3K inhibitor in combination with either the mTORC1/2 inhibitor sapanisertib or an AKT inhibitor, ipatasertib or afuresertib.
Collapse
Affiliation(s)
- Thomas S Dexheimer
- Molecular Pharmacology Laboratory, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, 1050 Boyles Street, Frederick, MD, 21702, USA.
| | - Zahra Davoudi
- Molecular Pharmacology Laboratory, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, 1050 Boyles Street, Frederick, MD, 21702, USA
| | - Nathan P Coussens
- Molecular Pharmacology Laboratory, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, 1050 Boyles Street, Frederick, MD, 21702, USA
| | - Thomas Silvers
- Molecular Pharmacology Laboratory, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, 1050 Boyles Street, Frederick, MD, 21702, USA
| | - Joel Morris
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Naoko Takebe
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Rabih Said
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jeffrey A Moscow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - James H Doroshow
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Beverly A Teicher
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
15
|
Zhang S, Hong HI, Mak VCY, Zhou Y, Lu Y, Zhuang G, Cheung LWT. Vertical inhibition of p110α/AKT and N-cadherin enhances treatment efficacy in PIK3CA-aberrated ovarian cancer cells. Mol Oncol 2025; 19:1132-1154. [PMID: 39543937 PMCID: PMC11977650 DOI: 10.1002/1878-0261.13761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 08/14/2024] [Accepted: 10/28/2024] [Indexed: 11/17/2024] Open
Abstract
Phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha [PIK3CA, encoding PI3Kalpha (also known as p110α)] is one of the most commonly aberrated genes in human cancers. In serous ovarian cancer, PIK3CA amplification is highly frequent but PIK3CA point mutation is rare. However, whether PIK3CA amplification and PIK3CA driver mutations have the same functional impact in the disease is unclear. Here, we report that both PIK3CA amplification and E545K mutation are tumorigenic. While the protein kinase B (AKT) signaling axis was activated in both E545K knock-in cells and PIK3CA-overexpressing cells, the mitogen-activated protein kinase 3/1 (ERK1/2) pathway was induced selectively by E545K mutation but not PIK3CA amplification. Intriguingly, AKT signaling in these PIK3CA-aberrated cells increased transcriptional coactivator YAP1 (YAP) Ser127 phosphorylation and thereby cytoplasmic YAP levels, which in turn increased cell migration through Ras-related C3 botulinum toxin substrate 1 (RAC1) activation. In addition to the altered YAP signaling, AKT upregulated N-cadherin expression, which also contributed to cell migration. Pharmacological inhibition of N-cadherin reduced cell migratory potential. Importantly, co-targeting N-cadherin and p110α/AKT caused additive reduction in cell migration in vitro and metastases formation in vivo. Together, this study reveals the molecular pathways driven by the PIK3CA aberrations and the exploitable vulnerabilities in PIK3CA-aberrated serous ovarian cancer cells.
Collapse
Affiliation(s)
- Shibo Zhang
- School of Biomedical Sciences, Li Ka Shing Faculty of MedicineThe University of Hong KongChina
- Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and TherapyGuangzhouChina
| | - Hei Ip Hong
- School of Biomedical Sciences, Li Ka Shing Faculty of MedicineThe University of Hong KongChina
| | - Victor C. Y. Mak
- School of Biomedical Sciences, Li Ka Shing Faculty of MedicineThe University of Hong KongChina
| | - Yuan Zhou
- School of Biomedical Sciences, Li Ka Shing Faculty of MedicineThe University of Hong KongChina
| | - Yiling Lu
- Division of Cancer Medicine, Department of Genomic MedicineUT MD Anderson Cancer CentreHoustonTXUSA
| | - Guanglei Zhuang
- State Key Laboratory of Systems Medicine for Cancer, Department of Obstetrics and Gynecology, Ren Ji Hospital, Shanghai Cancer InstituteShanghai Jiao Tong University School of MedicineChina
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji HospitalShanghai Jiao Tong University School of MedicineChina
| | - Lydia W. T. Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of MedicineThe University of Hong KongChina
| |
Collapse
|
16
|
Ondič O, Michalová K, Švajdler M, Presl J, Kosťun J, Hájková V, Martínek P, Michal M. Molecular substratification of endometrial carcinomas with no special molecular profile (NSMP) by using a limited NGS custom panel may facilitate effective patient selection for the PIK3CA-targeted therapy. Virchows Arch 2025; 486:827-832. [PMID: 39235514 PMCID: PMC12018627 DOI: 10.1007/s00428-024-03905-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/07/2024] [Accepted: 08/17/2024] [Indexed: 09/06/2024]
Abstract
Endometrial carcinomas (EC) of no special molecular profile (NSMP) represent the largest molecular category of EC, comprising a mixture of tumors with different histology and molecular profiles. These facts likely point to different tumor biology, clinical outcomes, and targeted therapy responses within this molecular category. The PIK3CA is currently the only targetable kinase oncoprotein directly implicated in EC carcinogenesis. Investigating a unique single-institution cohort, we attempted to stratify NSMP ECs based on the presence of the PIK3CA pathogenic mutation. Those cases were further analyzed for other well-established-associated oncogenic driver gene mutations. Histological and clinical variables were also correlated in each case. Altogether, 175 ECs were prospectively tested by a limited custom NGS panel containing ARID1A, BCOR, BRCA1, BRCA2, CTNNB1, KRAS, MLH1, MSH2, MSH6, NRAS, PIK3CA, PMS2, POLD1, POLE, PTEN,and TP53 genes. We identified 24 PIK3CA mutated cases in the group of 80 NSMP ECs, with another co-occurring mutation in at least one oncogenic driver gene (CTNNB1, PTEN, ARID1A, KRAS, BCOR, PMS2) in 19 cases. In conclusion, a limited NGS panel can effectively test EC tissue for specific pathogenetically relevant oncogene mutations. The NSMP EC category contains 30% of the PIK3CA mutated cases. Of those, 21% contain the PIK3CA mutation as a sole EC-associated oncogene mutation, while 79% harbor at least one more mutated gene. These findings may inform future healthcare planning and improve the effectiveness of EC patient selection for the PIK3CA-targeted therapy.
Collapse
Affiliation(s)
- Ondrej Ondič
- Department of Pathology, Medical Faculty in Pilsen, Charles University, Prague, Czech Republic.
- Molecular Genetics Department, Bioptická Laboratoř s.r.o, Pilsen, Czech Republic.
| | - Květoslava Michalová
- Department of Pathology, Medical Faculty in Pilsen, Charles University, Prague, Czech Republic
- Molecular Genetics Department, Bioptická Laboratoř s.r.o, Pilsen, Czech Republic
| | - Marián Švajdler
- Department of Pathology, Medical Faculty in Pilsen, Charles University, Prague, Czech Republic
- Molecular Genetics Department, Bioptická Laboratoř s.r.o, Pilsen, Czech Republic
| | - Jiří Presl
- Department of Gynecology and Obstetrics, Medical Faculty in Pilsen, Charles University, Prague, Czech Republic
| | - Jan Kosťun
- Department of Gynecology and Obstetrics, Medical Faculty in Pilsen, Charles University, Prague, Czech Republic
| | - Veronika Hájková
- Molecular Genetics Department, Bioptická Laboratoř s.r.o, Pilsen, Czech Republic
| | - Petr Martínek
- Molecular Genetics Department, Bioptická Laboratoř s.r.o, Pilsen, Czech Republic
| | - Michal Michal
- Department of Pathology, Medical Faculty in Pilsen, Charles University, Prague, Czech Republic
| |
Collapse
|
17
|
Sun J, Zhang Z, Xia B, Yao T, Ge F, Yan F. Overexpression of PIK3CG in Cancer Cells Promotes Lung Cancer Cell Migration and Metastasis Through Enhanced MMPs Expression and Neutrophil Recruitment and Activation. Biochem Genet 2025; 63:1647-1659. [PMID: 38602596 DOI: 10.1007/s10528-024-10788-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 03/16/2024] [Indexed: 04/12/2024]
Abstract
Metastasis is a major cause of death in lung cancer. The aim of this study is to analyze the role and mechanism of PI3K catalytic subunit gamma (PIK3CG, also known as p110γ) in lung cancer cell migration and metastasis. Knockdown (KD) and overexpression (OE) of PIK3CG expression in lung cancer cell lines A549 and H1299 in vitro cultured was achieved. Two PIK3CG-specific inhibitors, Eganelisib and CAY10505, were used to treat A549 and H1299 cells. An experimental lung metastasis mouse model was constructed using tail vein injection of LLC cells. Finally, a co-culture system was established using Transwell chambers. Compared with the NC group, the number of cells that completed migration and the expression levels of matrix metalloproteinases (MMPs) were significantly reduced in the KD group and Eganelisib and CAY10505 treatment groups, while the number of cells that migrated successfully and the expression levels of MMPs were significantly increased in the OE group. Lung tissues of mice injected with PIK3CG-stabilized overexpressed LLC cells showed more pronounced lung cancer growth, lung metastatic nodules, neutrophil infiltration and MMPs expression. Co-culture with neutrophils, soluble extracts of neutrophils and cathepsin G all promoted the migration of lung cancer cells. PIK3CG overexpression in tumor cells significantly promoted the migration and metastasis of lung cancer cell.
Collapse
Affiliation(s)
- Jinpeng Sun
- Department of General Surgery Ward, Cangzhou Hospital of Integrated TCM-WM, No. 31 Yellow River West Road, Canal District, Cangzhou, 061000, Hebei, China.
| | - Zhenshan Zhang
- Department of Medical Oncology, Cangzhou Hospital of Integrated TCM-WM, Cangzhou, 061000, Hebei, China
| | - Binghui Xia
- Department of General Surgery Ward, Cangzhou Hospital of Integrated TCM-WM, No. 31 Yellow River West Road, Canal District, Cangzhou, 061000, Hebei, China
| | - Tianyu Yao
- Department of Cardiology, Cangzhou Hospital of Integrated TCM-WM, Cangzhou, 061000, Hebei, China
| | - Fengyue Ge
- Department of Function Laboratory, Cangzhou Hospital of Integrated TCM-WM, Cangzhou, 061000, Hebei, China
| | - Fengmei Yan
- Department of Endoscopic Diagnosis and Treatment Center, Cangzhou Hospital of Integrated TCM-WM, Cangzhou, 061000, Hebei, China
| |
Collapse
|
18
|
Cao KY, Zhang D, Bai LB, Yan TM, Chen Y, Jiang YY, Jiang ZH. Targeting NUCKS1 with a fragment of tRNA Asn(GUU) of Chinese yew for the treatment of colorectal cancer. Noncoding RNA Res 2025; 11:38-47. [PMID: 39736854 PMCID: PMC11683283 DOI: 10.1016/j.ncrna.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/22/2024] [Accepted: 11/11/2024] [Indexed: 01/01/2025] Open
Abstract
Despite the discovery of numerous oncogenes in colorectal cancer (CRC), the development of associated drugs is limited, posing a significant challenge for CRC treatment. Identification of novel druggable targets is therefore crucial for the therapeutic development of CRC. Here, we report the first investigation on therapeutics targeting the potent oncogene NUCKS1 to suppress cancer progression. NUCKS1-orientated bioinformatics screening of NUCKS1 inhibitors from our library of tRNA fragments originated from medicinal plants identified tRF-T36, a 5' tRNA fragment of tRNAAsn(GUU) of Chinese yew (Taxus chinensis), exhibiting stronger inhibitory effects than taxol against CRC progression. Mechanistically, tRF-T36 binds directly to the 3' UTR of NUCKS1 mRNA to downregulate its expressions via RNAi pathway. High-throughput RNA sequencing indicated that the downregulated NUCKS1 induced by tRF-T36 further inhibits PI3K/Akt pathway, as verified by the significantly efficacy decrease of tRF-T36 mimic in co-treatment with 740Y-P, an agonist of PI3K/Akt pathway. Collectively, our findings emphasize the importance of NUCKS1 as a promising druggable target for CRC. Furthermore, the present study provides the first siRNA sequence, tRF-T36 mimic, as small RNA drug candidate, thereby shedding light on CRC therapeutics.
Collapse
Affiliation(s)
- Kai-Yue Cao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau SAR, China
| | - Da Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau SAR, China
| | - Long-Bo Bai
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau SAR, China
| | - Tong-Meng Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau SAR, China
| | - Yan Chen
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Yu-Yang Jiang
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Zhi-Hong Jiang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau SAR, China
| |
Collapse
|
19
|
Toklu A, Panse G, Jour G, Linos K, Cloutier JM, Dehner CA. Hybrid granular cell tumor/perineurioma: a report of two rare cases with PIK3CA mutations. Virchows Arch 2025:10.1007/s00428-025-04089-3. [PMID: 40159563 DOI: 10.1007/s00428-025-04089-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/27/2025] [Accepted: 03/22/2025] [Indexed: 04/02/2025]
Abstract
Hybrid peripheral nerve sheath tumors (PNSTs) are rare mesenchymal neoplasms with dual differentiation, most often combining two of the three main PNST types: schwannoma, neurofibroma, and perineurioma. Recognized by the WHO since 2013, these tumors can also exhibit fewer common combinations, such as hybrid granular cell tumor/perineurioma. We herein report two rare cases of hybrid granular cell tumor perineurioma with molecular analysis. Both tumors presented as dermal to subcutaneous, well-circumscribed lesions composed of a combination of spindled and granular cell components. By immunohistochemistry, the granular cells were positive for S100, SOX10, and CD68, while the perineurial cells were highlighted by EMA and GLUT1 stains. Subsequent molecular testing revealed pathogenic mutations involving PIK3CA in both cases. Our study expands on the clinical and pathologic spectrum and provides the first molecular data on these unusual neoplasms. Further, we provide a comprehensive review of the literature of all previously reported cases and briefly discuss relevant differential diagnoses and their molecular features.
Collapse
Affiliation(s)
- Ani Toklu
- Department of Pathology and Laboratory Medicine, Indiana University Hospital, Indianapolis, IN, 46202, USA
| | - Gauri Panse
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - George Jour
- Department of Pathology and Dermatology, New York School of Medicine, New York, NY, USA
| | - Konstantinos Linos
- Department of Pathology & Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jeffrey M Cloutier
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Carina A Dehner
- Department of Pathology and Laboratory Medicine, Indiana University Hospital, Indianapolis, IN, 46202, USA.
| |
Collapse
|
20
|
de Moraes FCA, Sano VKT, Pereira CRM, de Laia EA, Stecca C, Magalhães MCF, Tarantino P. Effects of AKT Inhibitors for PIK3CA/AKT1/PTEN-Altered Advanced or Metastatic Breast Cancer: A Meta-Analysis of Randomized Clinical Trials. Clin Breast Cancer 2025:S1526-8209(25)00079-5. [PMID: 40254500 DOI: 10.1016/j.clbc.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 03/03/2025] [Accepted: 03/16/2025] [Indexed: 04/22/2025]
Abstract
PURPOSE We aimed to answer the following question: How effective is the addition of AKT inhibitors to the treatment of advanced or metastatic breast cancer? METHODS We searched PubMed, Embase and Cochrane for randomized controlled trials (RCTs) that investigated AKT inhibitors for advanced or metastatic BC. We computed hazard-ratios (HRs) for binary endpoints. RESULTS A total of 5 RCTs were included in the meta-analysis, comprising 1,334 patients with BC. The use of AKT inhibitors demonstrated a significant improvement in OS (HR 0.70; 95% CI, 0.58-0.85; P < .001) and PFS (HR 0.6797; 95% CI, 0.5499-0.8403; P < .001) in the overall population. Within the PIK3CA/AKT1/PTEN-altered subgroup (n = 645), the OS rate also significantly favored AKT inhibitors over the control group (HR 0.62; 95% CI, 0.42-0.92; P = .019), as well as PFS (HR 0.5224; 95% CI, 0.3366-0.8105; P = .004). CONCLUSIONS Our findings suggest that the incorporation of AKT inhibitors holds promise for treating patients with advanced or metastatic PIK3CA/AKT1/PTEN-altered BC.
Collapse
Affiliation(s)
| | | | | | | | - Carlos Stecca
- Department of Medical Oncology, Mackenzie Evangelical University Hospital, Curitiba, Paraná, Brazil
| | | | - Paolo Tarantino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
21
|
Razzaq S, Fatima I, Moafian Z, Rahdar A, Fathi-Karkan S, Kharaba Z, Shirzad M, Khan A, Pandey S. Nanomedicine innovations in colon and rectal cancer: advances in targeted drug and gene delivery systems. Med Oncol 2025; 42:113. [PMID: 40097759 DOI: 10.1007/s12032-025-02670-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 03/04/2025] [Indexed: 03/19/2025]
Abstract
Nanotechnology has revolutionized cancer diagnostics and therapy, offering unprecedented possibilities to overcome the constraints of conventional treatments. This study provides a detailed overview of the current progress and difficulties in the creation of nanostructured materials, with a specific emphasis on their use in drug and gene delivery systems. The study examines tactics that attempt to improve the effectiveness and safety of chemotherapeutic drugs such as doxorubicin (Dox) by focusing on the potential of antibody-drug conjugates and functionalized nanoparticles. Moreover, it clarifies the challenges encountered in administering nanoparticles orally for gastrointestinal treatments, emphasizing the crucial physicochemical properties that affect their behavior in the gastrointestinal system. This study highlights the transformational potential of nanostructured materials in precision oncology by examining advanced breakthroughs such cell membrane-camouflaged nanoparticles and inorganic nanoparticles designed for gastrointestinal disorders. The text investigates the processes involved in the absorption of nanoparticles and their destruction in lysosomes, revealing the many methods in which enterocytes take up these particles. This study strongly supports the use of advanced nanoparticle-based methods to reduce the harmful effects on the whole body and improve the effectiveness of therapy, based on a thorough examination of current experiments on animals and humans. The main objective of this paper is to provide a fundamental comprehension that will stimulate more investigation and practical use in the field of cancer nanomedicine, advancing its boundaries.
Collapse
Affiliation(s)
- Sobia Razzaq
- School of Pharmacy, University of Management and Technology, Lahore, Punjab, Pakistan
| | - Iqra Fatima
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Zeinab Moafian
- Department of Chemistry and Biochemistry, University of Delaware, Newark, USA
| | - Abbas Rahdar
- Department of Physics, Faculty of Sciences, University of Zabol, Zabol, 538-98615, Iran.
| | - Sonia Fathi-Karkan
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 94531-55166, Iran.
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, 9414974877, Iran.
| | - Zelal Kharaba
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, UAE
| | - Maryam Shirzad
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ahmad Khan
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, 45320, Pakistan.
| | - Sadanand Pandey
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, 173229, Himachal Pradesh, India.
- Department of Chemistry, College of Natural Science, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk, 38541, Republic of Korea.
| |
Collapse
|
22
|
Fu H, Mo X, Ivanov AA. Decoding the functional impact of the cancer genome through protein-protein interactions. Nat Rev Cancer 2025; 25:189-208. [PMID: 39810024 DOI: 10.1038/s41568-024-00784-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/02/2024] [Indexed: 01/16/2025]
Abstract
Acquisition of genomic mutations enables cancer cells to gain fitness advantages under selective pressure and, ultimately, leads to oncogenic transformation. Interestingly, driver mutations, even within the same gene, can yield distinct phenotypes and clinical outcomes, necessitating a mutation-focused approach. Conversely, cellular functions are governed by molecular machines and signalling networks that are mostly controlled by protein-protein interactions (PPIs). The functional impact of individual genomic alterations could be transmitted through regulated nodes and hubs of PPIs. Oncogenic mutations may lead to modified residues of proteins, enabling interactions with other proteins that the wild-type protein does not typically interact with, or preventing interactions with proteins that the wild-type protein usually interacts with. This can result in the rewiring of molecular signalling cascades and the acquisition of an oncogenic phenotype. Here, we review the altered PPIs driven by oncogenic mutations, discuss technologies for monitoring PPIs and provide a functional analysis of mutation-directed PPIs. These driver mutation-enabled PPIs and mutation-perturbed PPIs present a new paradigm for the development of tumour-specific therapeutics. The intersection of cancer variants and altered PPI interfaces represents a new frontier for understanding oncogenic rewiring and developing tumour-selective therapeutic strategies.
Collapse
Affiliation(s)
- Haian Fu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Emory University, Atlanta, GA, USA.
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
| | - Xiulei Mo
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Emory University, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Andrey A Ivanov
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Emory University, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| |
Collapse
|
23
|
Wang Y, Rozen V, Zhao Y, Wang Z. Oncogenic activation of PI K3 CA in cancers: Emerging targeted therapies in precision oncology. Genes Dis 2025; 12:101430. [PMID: 39717717 PMCID: PMC11665392 DOI: 10.1016/j.gendis.2024.101430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/04/2024] [Accepted: 08/25/2024] [Indexed: 12/25/2024] Open
Abstract
Phosphoinositide 3-kinases (PI3Ks) are heterodimers consisting of a p110 catalytic subunit and a p85 regulatory subunit. The PIK3CA gene, which encodes the p110α, is the most frequently mutated oncogene in cancer. Oncogenic PIK3CA mutations activate the PI3K pathway, promote tumor initiation and development, and mediate resistance to anti-tumor treatments, making the mutant p110α an excellent target for cancer therapy. PIK3CA mutations occur in two hotspot regions: one in the helical domain and the other in the kinase domain. The PIK3CA helical and kinase domain mutations exert their oncogenic function through distinct mechanisms. For example, helical domain mutations of p110α gained direct interaction with insulin receptor substrate 1 (IRS-1) to activate the downstream signaling pathways. Moreover, p85β proteins disassociate from helical domain mutant p110α, translocate into the nucleus, and stabilize enhancer of zeste homolog 1/2 (EZH1/2). Due to the fundamental role of PI3Kα in tumor initiation and development, PI3Kα-specific inhibitors, represented by FDA-approved alpelisib, have developed rapidly in recent decades. However, side effects, including on-target side effects such as hyperglycemia, restrict the maximum dose and thus clinical efficacy of alpelisib. Therefore, developing p110α mutant-specific inhibitors to circumvent on-target side effects becomes a new direction for targeting PIK3CA mutant cancers. In this review, we briefly introduce the function of the PI3K pathway and discuss how PIK3CA mutations rewire cell signaling, metabolism, and tumor microenvironment, as well as therapeutic strategies under development to treat patients with tumors harboring a PIK3CA mutation.
Collapse
Affiliation(s)
- Yuxiang Wang
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Valery Rozen
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Yiqing Zhao
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Zhenghe Wang
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
24
|
Li X, Zhang X, Yin S, Nie J. Challenges and prospects in HER2-positive breast cancer-targeted therapy. Crit Rev Oncol Hematol 2025; 207:104624. [PMID: 39826885 DOI: 10.1016/j.critrevonc.2025.104624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 12/29/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025] Open
Abstract
Breast cancer remains the most prevalent malignancy among women globally and ranks as the leading cause of cancer-related mortality in this demographic. Approximately 13 %-15 % of all breast cancer cases are classified as HER2-positive, a subtype associated with a particularly unfavorable prognosis. A large number of patients with HER2-positive breast cancer continue to face disease progression after receiving standardized treatment. Given these challenges, a thorough exploration into the mechanisms underlying drug resistance in HER2-targeted therapy is imperative. This review focuses on the factors related to drug resistance in HER2-targeted therapy, including tumor heterogeneity, antibody-binding efficacy, variations in the tumor microenvironment, and abnormalities in signal activation and transmission. Additionally, corresponding strategies to counteract these resistance mechanisms are discussed, to advance therapeutic efficacy and clinical benefits in the management of HER2-positive breast cancer.
Collapse
Affiliation(s)
- Xiyin Li
- Department of Breast Cancer, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, the Third Affiliated Hospital, Kunming Medical University, 519 Kunzhou Road, Kunming, Yunnan 650118, China.
| | - Xueying Zhang
- Department of Breast Cancer, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, the Third Affiliated Hospital, Kunming Medical University, 519 Kunzhou Road, Kunming, Yunnan 650118, China.
| | - Saige Yin
- Department of Anatomy and Histology and Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan 650118, China.
| | - Jianyun Nie
- Department of Breast Cancer, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, the Third Affiliated Hospital, Kunming Medical University, 519 Kunzhou Road, Kunming, Yunnan 650118, China.
| |
Collapse
|
25
|
Szostakowska-Rodzos M, Grzybowska EA, Mysliwy I, Zub R, Jagiello-Gruszfeld A, Rubach M, Konieczna A, Fabisiewicz A. The Combined Assessment of CTC and ESR1 Status in Liquid Biopsy Samples Enhances the Clinical Value of Prediction in Metastatic Breast Cancer. Int J Mol Sci 2025; 26:2038. [PMID: 40076662 PMCID: PMC11900918 DOI: 10.3390/ijms26052038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Monitoring of metastatic breast cancer (mBC) is an important issue in the clinical management of patients. Liquid biopsy has become a non-invasive method for detecting and monitoring cancer in body fluids. The presence of circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) in peripheral blood indicates poor prognosis and may contribute to early detection of progression, but assessment of these levels is still not routine clinical management. The main objective of this study was to estimate the frequency and clinical value of the ESR1 and PIK3CA mutations identified in circulating free DNA (cfDNA.) The second goal was to evaluate whether simultaneous evaluation of CTCs and mutation status in cfDNA increases the prognostic value of liquid biopsy. The results of the analysis of the CTC number and ESR1 and PIK3CA mutations in blood collected from 179 patients with metastatic breast cancer show that ESR1 mutations are more frequent in patients with advanced luminal breast cancer regardless of the type of the treatment. ESR1 mutations appear primarily during progression, as no mutations were found in primary tumor samples. The main conclusion of the study is that combined assessment of CTCs and ESR1 status in liquid biopsy may improve the prognostic value of liquid biopsy.
Collapse
Affiliation(s)
- Malgorzata Szostakowska-Rodzos
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland; (M.S.-R.); (I.M.)
| | - Ewa A. Grzybowska
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland; (M.S.-R.); (I.M.)
| | - Izabella Mysliwy
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland; (M.S.-R.); (I.M.)
| | - Renata Zub
- Cancer Molecular and Genetic Diagnostics Department, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland;
| | | | - Maryna Rubach
- Cancer Chemotherapy Day Unit, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland;
| | - Aleksandra Konieczna
- Department of Breast Cancer and Reconstructive Surgery, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland;
| | - Anna Fabisiewicz
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland; (M.S.-R.); (I.M.)
| |
Collapse
|
26
|
Zhang HL, Lin Z, Zhang Y. Developments in research and commercialization of PI3K and AKT targets: a patent-based landscape. Pharm Pat Anal 2025:1-8. [PMID: 39993965 DOI: 10.1080/20468954.2025.2470102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 02/18/2025] [Indexed: 02/26/2025]
Abstract
PI3K and AKT signaling pathway has been linked to the pathophysiology of various diseases. This pathway has emerged as a crucial therapeutic strategy for cancer and other diseases. To better understand recent development of PI3K and AKT, a patent-based landscape study was performed. The results shows that both PI3K and AKT targets have shown prolific patent filings over the past 20 years. This study is the first to depict the therapeutic applications of both PI3K and AKT targets based on a patent big data analysis. Ten key therapeutic applications were identified, with over 77% of patents related to anti-cancer therapy for both PI3K and AKT targets. Additionally, our findings show that combination therapy is a distinguishing feature for drugs targeting both PI3K and AKT. The average time from patent application to drug approval for PI3K target drugs is 8.8 years. PI3K target drugs obtain market approval faster compared to AKT drugs. Approximately, 2 years of patent term extension could be obtained if the time from the patent application date to the drug approval date is less than 10 years.
Collapse
Affiliation(s)
- Hai-Long Zhang
- Central International Intellectual Property (Baotou) Co. Ltd, Baotou, China
| | - Zhaochen Lin
- Hydrogen Medicine Research Centre, The Affiliated Taian City Central Hospital of Qingdao University, Tai'an, Shandong, China
| | - Ying Zhang
- Pharmacy Intravenous Admixture Services, The Affiliated Taian City Central Hospital of Qingdao University, Tai'an, Shandong, China
| |
Collapse
|
27
|
Jani V, Sonavane U, Sawant S. Understanding the conformational dynamics of PI3Kα due to helical domain mutations: insights from Markov state model analysis. Mol Divers 2025:10.1007/s11030-025-11138-1. [PMID: 39982680 DOI: 10.1007/s11030-025-11138-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/12/2025] [Indexed: 02/22/2025]
Abstract
Phosphoinositide 3-kinases (PI3Ks) phosphorylate phosphoinositides on the membrane, which act as secondary signals for various cellular processes. PI3Kα, a heterodimer of the p110α catalytic subunit and the p85α regulatory subunit, is activated by growth factor receptors or mutations. Among these mutations, E545K present in the helical domain is strongly associated with cancer, and is known to disrupt interactions between the regulatory and catalytic subunits, leading to its constitutive activation. However, while the mutation's role in disrupting autoinhibition is well documented, the molecular mechanisms linking this mutation in the helical domain to the structural changes in the kinase domain remain poorly understood. This study aims to understand the conformational events triggered by the E545K mutation, elucidate how these changes propagate from the helical domain to the kinase domain, and identify crucial residues involved in the activation process. Molecular dynamics (MD) simulations combined with Markov state modeling (MSM) were employed to explore the conformational landscapes of both the wild-type and mutant systems. Structural and energetic analyses, including Molecular Mechanics Poisson-Boltzmann Surface Area (MM-PBSA) calculations, revealed that the E545K mutation significantly reduces the binding affinity between the regulatory and catalytic subunits. The mutation was found to induce a sliding motion of the regulatory subunit along the catalytic subunit, leading to the disruption of key salt-bridges between these domains. This disruption releases the inhibitory effect of the regulatory subunit, resulting in increased domain motion, particularly in the adaptor-binding domain (ABD). Enhanced flexibility in the ABD, helical, and C2 domains facilitates the rearrangement of the two lobes of kinase domain, thereby promoting activation. Additionally, the mutation appears to enhance PI3Kα's membrane affinity via the Ras-binding domain (RBD). Network analysis helped to identify key residues that may involve in allosteric signaling pathways, providing insights into the communication between domains. Druggable pockets in the metastable states were predicted followed by its docking with a PI3K inhibitor library. Docking studies revealed the crucial residues that may be participating in inhibitor binding. The identification of residues and regions involved in activation mechanisms using MSM helped to reveal the conformational events and the knowledge on probable allosteric pockets, which may be helpful in designing better therapeutics.
Collapse
Affiliation(s)
- Vinod Jani
- HPC-M&BA Group, Centre for Development of Advanced Computing, Pune, 411008, India
- Bioinformatics Centre, Savitribai Phule Pune University, Pune, 411007, India
| | - Uddhavesh Sonavane
- HPC-M&BA Group, Centre for Development of Advanced Computing, Pune, 411008, India.
| | - Sangeeta Sawant
- Bioinformatics Centre, Savitribai Phule Pune University, Pune, 411007, India
| |
Collapse
|
28
|
Hedger G, Yen HY. The Influence of Phosphoinositide Lipids in the Molecular Biology of Membrane Proteins: Recent Insights from Simulations. J Mol Biol 2025; 437:168937. [PMID: 39793883 PMCID: PMC7617384 DOI: 10.1016/j.jmb.2025.168937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/29/2024] [Accepted: 01/06/2025] [Indexed: 01/13/2025]
Abstract
The phosphoinositide family of membrane lipids play diverse and critical roles in eukaryotic molecular biology. Much of this biological activity derives from interactions of phosphoinositide lipids with integral and peripheral membrane proteins, leading to modulation of protein structure, function, and cellular distribution. Since the discovery of phosphoinositides in the 1940s, combined molecular biology, biophysical, and structural approaches have made enormous progress in untangling this vast and diverse cellular network of interactions. More recently, in silico approaches such as molecular dynamics simulations have proven to be an asset in prospectively identifying, characterising, explaining the structural basis of these interactions, and in the best cases providing atomic level testable hypotheses on how such interactions control the function of a given membrane protein. This review details a number of recent seminal discoveries in phosphoinositide biology, enabled by advanced biomolecular simulation, and its integration with molecular biology, biophysical, and structural biology approaches. The results of the simulation studies agree well with experimental work, and in a number of notable cases have arrived at the key conclusion several years in advance of the experimental structures. SUMMARY: Hedger and Yen review developments in simulations of phosphoinositides and membrane proteins.
Collapse
Affiliation(s)
- George Hedger
- Department of Life Sciences, Sir Ernst Chain Building, Imperial College London, London, SW7 2AZ, UK.
| | - Hsin-Yung Yen
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan; Department of Chemistry, University of Oxford, South Parks Road, Oxford, OX1 3QZ, UK
| |
Collapse
|
29
|
Xu J, You Z, Zhu Z, Liu M, Zhang Z, Xu P, Dong J, Huang Y, Wang C, Qin H. Integrative analysis of m7G methylation-associated genes prognostic signature with immunotherapy and identification of LARP1 as a key oncogene in head and neck squamous cell carcinoma. Front Immunol 2025; 16:1520070. [PMID: 40018039 PMCID: PMC11864954 DOI: 10.3389/fimmu.2025.1520070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/21/2025] [Indexed: 03/01/2025] Open
Abstract
Background N7-methylguanosine (m7G) methylation is an RNA modification associated with cancer progression, but its specific role in head and neck squamous cell carcinoma (HNSCC) remains unclear. Methods This study analyzed the differential expression of m7G-related genes (m7GRGs) in HNSCC using the TCGA-HNSCC dataset, identifying key pathways associated with the cell cycle, DNA replication, and focal adhesion. A LASSO-Cox regression model was constructed based on four m7GRGs (EIF3D, EIF1, LARP1, and METTL1) and validated with GEO datasets and clinical samples. Further validation of gene upregulation in HNSCC tissues was conducted using RT-qPCR and immunohistochemistry, while the role of LARP1 in HNSCC cells was assessed via knockout experiments. Results The constructed model demonstrated strong predictive performance, with the risk score significantly correlating with prognosis, immune infiltration, and drug sensitivity. An external dataset and clinical specimens further confirmed the model's predictive accuracy for immunotherapy response. Additionally, two regulatory axes-LINC00707/hsa-miR-30b-5p/LARP1 and SNHG16/hsa-miR-30b-5p/LARP1-were identified. LARP1 knockout experiments revealed that suppressing LARP1 markedly inhibited HNSCC cell proliferation, migration, and invasion. Conclusion The m7GRG-based prognostic model developed in this study holds strong clinical potential for predicting prognosis and therapeutic responses in HNSCC. The identification of LARP1 and its related regulatory pathways offers new avenues for targeted therapy in HNSCC.
Collapse
Affiliation(s)
- Juan Xu
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Zihao You
- Anhui Medical University, Hefei, China
| | | | - Min Liu
- Emergency Department, Peking University Shenzhen Hospital, Shenzhen, China
| | - Zheng Zhang
- Stomatological Center, Peking University Shenzhen Hospital, Shenzhen, China
| | - Panpan Xu
- Department of Otolaryngology Head and Neck Surgery, Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Juanjuan Dong
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Yuting Huang
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Chao Wang
- Department of Oncology, Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Haotian Qin
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
30
|
Weber-Levine C, Rakovec M, Jiang K, Kalluri A, Raj D, Parker M, Materi J, Sepehri S, Ferrés A, Schreck KC, Aldecoa I, Lucas CHG, Redmond KJ, Holdhoff M, Sair HI, Weingart JD, Brem H, González Sánchez J, Ye X, Bettegowda C, Rincon-Torroella J. Genomic Alterations in Molecularly Defined Oligodendrogliomas. Neurosurgery 2025; 96:328-337. [PMID: 39007559 DOI: 10.1227/neu.0000000000003078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/26/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Oligodendrogliomas are defined by IDH1/2 mutation and codeletion of chromosome arms 1p/19q. Although previous studies identified CIC , FUBP1 , and TERTp as frequently altered in oligodendrogliomas, the clinical relevance of these molecular signatures is unclear. Moreover, previous studies predominantly used research panels that are not readily available to providers and patients. Accordingly, we explore genomic alterations in molecularly defined oligodendrogliomas using clinically standardized next-generation sequencing (NGS) panels. METHODS A retrospective single-center study evaluated adults with pathologically confirmed IDH -mutant, 1p/19q-codeleted oligodendrogliomas diagnosed between 2005 and 2021. Genetic data from formalin-fixed, paraffin-embedded specimens were analyzed with the NGS Solid Tumor Panel at the Johns Hopkins Medical Laboratories, which tests more than 400 cancer-related genes. Kaplan-Meier plots and log-rank tests compared progression-free survival (PFS) and overall survival by variant status. χ 2 tests, t -tests, and Wilcoxon rank-sum tests were used to compare clinical characteristics between genomic variant status in the 10 most frequently altered genes. RESULTS Two hundred and seventy-seven patients with molecularly defined oligodendrogliomas were identified, of which 95 patients had available NGS reports. Ten genes had 9 or more patients with a genomic alteration, with CIC , FUBP1 , and TERTp being the most frequently altered genes (n = 60, 23, and 22, respectively). Kaplan-Meier curves showed that most genes were not associated with differences in PFS or overall survival. At earlier time points (PFS <100 months), CIC alterations conferred a reduction in PFS in patients ( P = .038). CONCLUSION Our study confirms the elevated frequency of CIC , FUBP1 , and TERTp alterations in molecularly defined oligodendrogliomas and suggests a potential relationship of CIC alteration to PFS at earlier time points. Understanding these genomic variants may inform prognosis or therapeutic recommendations as NGS becomes routine.
Collapse
Affiliation(s)
- Carly Weber-Levine
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore , Maryland , USA
| | - Maureen Rakovec
- Department of Neurosurgery, University of Maryland Medical Center, Baltimore , Maryland , USA
| | - Kelly Jiang
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore , Maryland , USA
| | - Anita Kalluri
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore , Maryland , USA
| | - Divyaansh Raj
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore , Maryland , USA
| | - Megan Parker
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore , Maryland , USA
| | - Joshua Materi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore , Maryland , USA
| | - Sadra Sepehri
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore , Maryland , USA
| | - Abel Ferrés
- Department of Neurosurgery, Hospital Clínic de Barcelona, Barcelona , Spain
| | - Karisa C Schreck
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore , Maryland , USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore , Maryland , USA
| | - Iban Aldecoa
- Department of Pathology, Biomedical Diagnostic Center (CDB), Hospital Clínic de Barcelona, University of Barcelona, Barcelona , Spain
- Neurological Tissue Bank of the Biobank Hospital Clinic Barcelona-FCRB/IDIBAPS, Barcelona , Spain
| | - Calixto-Hope G Lucas
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore , Maryland , USA
| | - Kristin J Redmond
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore , Maryland , USA
| | - Matthias Holdhoff
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore , Maryland , USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore , Maryland , USA
| | - Haris I Sair
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore , Maryland , USA
- The Malone Center for Engineering in Healthcare, Whiting School of Engineering, Johns Hopkins University, Baltimore , Maryland , USA
| | - Jon D Weingart
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore , Maryland , USA
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore , Maryland , USA
| | - Josep González Sánchez
- Department of Neurosurgery, Hospital Clínic de Barcelona, Barcelona , Spain
- Laboratory of Experimental Oncological Neurosurgery, Neurosurgery Service, Hospital Clinic de Barcelona, Barcelona , Spain
| | - Xiaobu Ye
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore , Maryland , USA
| | - Chetan Bettegowda
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore , Maryland , USA
| | - Jordina Rincon-Torroella
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore , Maryland , USA
- Laboratory of Experimental Oncological Neurosurgery, Neurosurgery Service, Hospital Clinic de Barcelona, Barcelona , Spain
- Programa de Doctorat de Medicina i Recerca Translacional, Universitat de Barcelona, Barcelona , Spain
| |
Collapse
|
31
|
Ling S, Dexter A, Race AM, Sharma S, Hamm G, Polanska UM, Marshall JF, Takats Z, Brindle K, Yuneva MO, Poulogiannis G, Campbell AD, Sansom OJ, Goodwin RJA, Bunch J, Barry ST. Use of metabolic imaging to monitor heterogeneity of tumour response following therapeutic mTORC1/2 pathway inhibition. Dis Model Mech 2025; 18:DMM050804. [PMID: 40019006 PMCID: PMC11892681 DOI: 10.1242/dmm.050804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 12/16/2024] [Indexed: 03/01/2025] Open
Abstract
The PI3K-mTOR-AKT pathway regulates tumour proliferation, gene expression and metabolism, but pathway inhibition induces heterogeneous feedback reactivation, limiting anti-tumour responses. Measuring heterogeneity of pathway inhibition in tissues using protein biomarker phosphorylation or location is challenging. An integrated multi-modal imaging workflow was developed to assess the heterogeneity of AZD2014 (mTORC1/2 inhibitor) response in a PTEN-null renal cancer model. Spatial responses of metabolite biomarkers were analysed by mass spectrometry imaging (MSI). Control and treated tumours were classified according to metabolite-defined regions enriched in control versus AZD2014-treated tumours, respectively. Noticeably, AZD2014-treated tumours retained regions similar to regions dominant in untreated tumours. Imaging mass cytometry analysis of protein biomarkers in 'control-like' regions following AZD2014 treatment showed reduced phospho-S6, indicating suppression, but retained high expression of the glucose transporter GLUT1. Increasing PI3K-AKT inhibition by combining with AZD8186 (PI3Kβ inhibitor) further decreased the control-like metabolic signature, showing PI3K-dependent resistance. This demonstrates that MSI-based workflows yield novel insights into the pharmacodynamic effects of mTORC1/2 inhibition in tumours, which classical biomarkers do not resolve. Coupling these workflows with spatial-omics approaches can deliver greater insights into heterogeneity of treatment response.
Collapse
Affiliation(s)
- Stephanie Ling
- Imaging and Data Analytics, AstraZeneca, Cambridge CB2 0AA, UK
| | - Alex Dexter
- National Physical Laboratory, Teddington TW11 0LA, UK
| | - Alan M. Race
- National Physical Laboratory, Teddington TW11 0LA, UK
| | - Shreya Sharma
- National Physical Laboratory, Teddington TW11 0LA, UK
| | - Gregory Hamm
- Imaging and Data Analytics, AstraZeneca, Cambridge CB2 0AA, UK
| | | | | | | | - Zoltan Takats
- Imperial College London, London SW7 2AZ, UK
- The Rosalind Franklin Institute, Harwell Campus, Didcot OX11 0QS, UK
| | - Kevin Brindle
- The Rosalind Franklin Institute, Harwell Campus, Didcot OX11 0QS, UK
- CRUK Cambridge Institute, Cambridge CB2 0RE, UK
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Qiu S, Zong Z, He K. Atezolizumab and Bevacizumab prior to liver transplantation in hepatic angiosarcoma mimicking hepatocellular carcinoma. Immunotherapy 2025; 17:175-178. [PMID: 40083292 PMCID: PMC11951687 DOI: 10.1080/1750743x.2025.2478811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/10/2025] [Indexed: 03/16/2025] Open
Abstract
Hepatic angiosarcoma is a rare yet aggressive malignancy, which is prone to misdiagnosis due to the lack of specific molecular and radiological characteristics. The treatment regimens remain controversial and disappointing. Randomized clinical trials are limited due to the rarity of this specific aggressive malignancy. Independent case reports or series can provide relevant references for treatment. We reported the very first hepatic angiosarcoma received Atezolizumab and Bevacizumab as a first-line treatment prior to liver transplantation, which resulted in a partial pathological response under specific molecular mutations. This case illustrates the potential role of immune checkpoint inhibitor combined with anti-angiogenic therapy as an off-label treatment option warranting further investigation.
Collapse
Affiliation(s)
- Siqi Qiu
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhipeng Zong
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kang He
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
33
|
Tsai YF, Lai JI, Liu CY, Hsi CN, Hsu CY, Huang CC, Feng CJ, Lin YS, Chao TC, Chiu JH, Tseng LM. Correlation Between PIK3R1 Expression and Cell Growth in Human Breast Cancer Cell Line BT-474 and Clinical Outcomes. World J Oncol 2025; 16:131-141. [PMID: 39850525 PMCID: PMC11750754 DOI: 10.14740/wjon1986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/16/2024] [Indexed: 01/25/2025] Open
Abstract
Background While mutations in the PIK3CA gene play important roles in human breast carcinogenesis, PIK3R1 gene alterations are recognized as actionable mutations for clinical cancer treatment. We aimed to elucidate the role of PIK3R1 in cell proliferation on breast carcinoma and to correlate the PIK3R1 expression with patients' outcome using human tumor tissue arrays. Methods Using human BT-474 (estrogen receptor (ER)+/human epidermal growth factor receptor 2 (HER2)-high) breast carcinoma cell line as in vitro model, the role of PIK3R1 in cell proliferation was elucidated by knock-down of the PIK3R1 gene (ΔPIK3R1) in this cell line. Between January 2000 to December 2015, the records of a cohort of 440 patients in our hospital were retrospectively reviewed, including patients' survival. The correlations between PIK3R1 expression and patient prognosis, such as overall survival (OS) and disease-free survival (DFS), were elucidated by human breast cancer tumor tissue array immunostaining. Results After the PIK3R1 gene was silenced in the BT-474 line, there was an increased cell number and a decrease in the G0G1-fraction, and increased S-fraction and the S+G2M-fraction for the ΔPIK3R1-BT-474 cell line, as compared to their cell wild type (WT) line. Western blot analysis showed that decreased PIK3R1 protein levels were accompanied by an increase of the p-AKT and p-mTOR proteins in the ΔPIK3R1-BT-474 cell line, compared to the equivalent WT line. Using a human tumor tissue array, patients with high-expressed PIK3R1 protein had better outcomes in terms of DFS and OS, compared to those with low-expressed PIK3R1 protein, when breast cancer was at an early stage (stage I/II), but not across all stages of breast cancer in human patients. Conclusions We concluded that downregulated PIK3R1 in BT-474 cells resulted in an increased cell growth and upregulated AKT-mTOR signaling. Clinically, the high-expressed PIK3R1 protein in tumors correlates positively with patients' outcome in stage I and II breast cancer.
Collapse
Affiliation(s)
- Yi-Fang Tsai
- Comprehensive Breast Health Center, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Division of Breast Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112201, Taiwan
| | - Jiun-I Lai
- Comprehensive Breast Health Center, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112201, Taiwan
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Chun-Yu Liu
- Comprehensive Breast Health Center, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112201, Taiwan
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Chieh-Ning Hsi
- Comprehensive Breast Health Center, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Division of Breast Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Chih-Yi Hsu
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112201, Taiwan
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Chi-Cheng Huang
- Comprehensive Breast Health Center, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Division of Breast Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei 100233, Taiwan
| | - Chin-Jung Feng
- Comprehensive Breast Health Center, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112201, Taiwan
- Division of Plastic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Yen-Shu Lin
- Comprehensive Breast Health Center, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Division of Breast Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112201, Taiwan
| | - Ta-Chung Chao
- Comprehensive Breast Health Center, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112201, Taiwan
- Division of Cancer Prevention, Department of Oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Jen-Hwey Chiu
- Comprehensive Breast Health Center, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Division of Breast Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Division of General Surgery, Department of Surgery, Cheng-Hsin General Hospital, Taipei 112401, Taiwan
| | - Ling-Ming Tseng
- Comprehensive Breast Health Center, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Division of Breast Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112201, Taiwan
- These authors contributed equally to this work
| |
Collapse
|
34
|
Li J, Xue J, Liu T, Feng Y, Xu N, Huang J, Yin Y, Zhang J, Mou H, Shentu J, Bao H, Xu Z, Xu Z. Phase Ib study of the oral PI3Kδ inhibitor linperlisib in patients with advanced solid tumors. Int J Clin Oncol 2025; 30:241-251. [PMID: 39538003 PMCID: PMC11785675 DOI: 10.1007/s10147-024-02657-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Patients with advanced solid tumors have a suboptimal prognosis. This study investigated the safety and feasibility of linperlisib, a selective phosphatidylinositol 3-kinase delta isoform (PI3Kδ) inhibitor, for treating patients with advanced solid tumors. METHODS In this phase Ib, single-arm, open-label, multi-center clinical trial, patients with histologically confirmed advanced solid tumors from eight centers in China were enrolled to receive oral linperlisib (80 mg/day). The primary endpoint was safety. RESULTS Between August 2019 and June 2022, 94 patients were enrolled in the trial and received the study treatment. The most common (≥ 20%) treatment emergent adverse events (TEAEs) of all grades irrespective of causality were increased aspartate aminotransferase (AST) (26.6%), proteinuria (26.6%), decreased appetite (25.5%), increased alanine aminotransferase (ALT) (22.3%), weight loss (21.3%), and anemia (21.3%). The most common grade ≥ 3 TEAEs were diarrhea (4.3%), increased AST (3.2%), increased ALT (3.2%), neutropenia (3.2%), anemia (3.2%), increased blood alkaline phosphatase (3.2%). The objective response rate (ORR) was 1.1% (95% confidence interval [CI] 0.0-5.8), and the disease control rate (DCR) was 37.2% (95% CI 27.5-47.8). As of the data cutoff, the median follow-up time was 4.2 months (95% CI 2.8-6.9). The median progression-free survival (PFS) was 1.85 months (95% CI 1.79-1.88). The median overall survival (OS) was not reached. CONCLUSION Linperlisib showed an acceptable safety profile and preliminary clinical benefit in patients with a range of advanced solid tumors. Further studies of linperlisib safety and efficacy are warranted.
Collapse
Affiliation(s)
- Jin Li
- Department of Oncology, East Hospital Affiliated to Tongji University, No. 150 Jimo Road, Pudong New Area, Shanghai, 200120, China.
| | - Junli Xue
- Department of Oncology, East Hospital Affiliated to Tongji University, No. 150 Jimo Road, Pudong New Area, Shanghai, 200120, China
| | - Tianshu Liu
- Department of Oncology, Zhongshan Hospital Affiliated to Fudan University, Shanghai, 200032, China
| | - Yi Feng
- Department of Oncology, Zhongshan Hospital Affiliated to Fudan University, Shanghai, 200032, China
| | - Nong Xu
- Department of Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, 310003, China
| | - Jianjin Huang
- Department of Oncology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, 310009, China
| | - Yongmei Yin
- Department of Oncology, Jiangsu Provincial People's Hospital, Nanjing, 210029, China
| | - Jun Zhang
- Department of Oncology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Haibo Mou
- Department of Oncology, Zhejiang Shulan Hospital, Hangzhou, 310022, China
| | - Jiangzhong Shentu
- Department of Pharmacology, The First Affiliated Hospital of Zhejiang University, Hangzhou, 310003, China
| | - Hanying Bao
- Shanghai Yingli Pharmaceutical Co., Ltd., Shanghai, 201210, China
| | - Zusheng Xu
- Shanghai Yingli Pharmaceutical Co., Ltd., Shanghai, 201210, China
| | - Zuhong Xu
- Shanghai Yingli Pharmaceutical Co., Ltd., Shanghai, 201210, China
| |
Collapse
|
35
|
Andreoti TAA, Maiolo M, Tuleja A, Döring Y, Schaller A, Vassella E, Boon LM, Baumgartner I, Bernhard SM, Zweier C, Vikkula M, Rössler J. Non-Hotspot PIK3CA Variants Have Higher Variant Allele Frequency and are More Common in Syndromic Vascular Malformations. Am J Med Genet A 2025; 197:e63883. [PMID: 39376044 DOI: 10.1002/ajmg.a.63883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/29/2024] [Accepted: 09/06/2024] [Indexed: 10/09/2024]
Abstract
PIK3CA variants are known to cause vascular malformations. We were interested in studying the phenotypic spectrum, the location within the PIK3CA gene, and the variant allele frequency (VAF) of somatic PI3KCA variants in vascular malformations. Clinical data of consecutive patients with extracranial/extraspinal vascular malformations were collected in the context of the VASCOM cohort (2008-2022, n = 558). Starting October 2020, biopsy samples were tested with the TSO500 gene panel (Illumina). All consenting patients with PIK3CA variants were included in this study. Eighty-nine patients had available genetic results by June 2022. PIK3CA variants (n = 25) were found in 16 simple/combined (nonsyndromic) vascular malformations and in nine vascular malformations associated with other anomalies (syndromic). Four hotspot variants in exons 9 and 20 (c.1624G>A, c.1633G>A, c.3140A>G, c.3140A>T) were identified in 16/25 patients (VAF 0.9%-9.7%). Six non-hotspot variants (c.328_330del, c.323_337del, c.353G>A, c.1258T>C, c.3132T>A, c.3195_3203delinsT) were detected in nine patients (VAF 3.6%-31.7%). Non-hotspot variants were more frequent in syndromic than nonsyndromic vascular malformations (p = 0.0034) and exhibited a higher VAF than hotspot variants (p = 0.0253). Our study contributes to the growing body of knowledge of the genetic background in vascular malformations. Further studies will enrich the ever-growing list of pathogenic PIK3CA variants associated with vascular malformations.
Collapse
Affiliation(s)
- Themis-Areti A Andreoti
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Inselspital - University Hospital of Bern, University of Bern, Bern, Switzerland
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland
| | - Massimo Maiolo
- Division of Angiology, Swiss Cardiovascular Center, Inselspital - University Hospital of Bern, University of Bern, Bern, Switzerland
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Aleksandra Tuleja
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland
- Division of Angiology, Swiss Cardiovascular Center, Inselspital - University Hospital of Bern, University of Bern, Bern, Switzerland
| | - Yvonne Döring
- Division of Angiology, Swiss Cardiovascular Center, Inselspital - University Hospital of Bern, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - André Schaller
- Department of Human Genetics, Inselspital - University Hospital of Bern, University of Bern, Bern, Switzerland
| | - Erik Vassella
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Laurence M Boon
- Center for Vascular Anomalies, Cliniques Universitaires Saint-Luc, Brussels, Belgium
- VASCERN (European Network of Rare Vascular Diseases) Reference Center, VASCA Working Group, Brussels, Belgium
| | - Iris Baumgartner
- Vasc Alliance AG, Bern, Switzerland
- Bern Center for Vascular Medicine and Interventions, Bern, Switzerland
| | - Sarah M Bernhard
- Division of Angiology, Swiss Cardiovascular Center, Inselspital - University Hospital of Bern, University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Vascular Center, Bienna Hospital Center, Bienna, Switzerland
| | - Christiane Zweier
- Department of Human Genetics, Inselspital - University Hospital of Bern, University of Bern, Bern, Switzerland
| | - Miikka Vikkula
- Center for Vascular Anomalies, Cliniques Universitaires Saint-Luc, Brussels, Belgium
- VASCERN (European Network of Rare Vascular Diseases) Reference Center, VASCA Working Group, Brussels, Belgium
- Department of Human Genetics, Laboratory of Human Molecular Genetics, de Duve Institute, Catholic University of Louvain, Brussels, Belgium
| | - Jochen Rössler
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Inselspital - University Hospital of Bern, University of Bern, Bern, Switzerland
- Department of Vascular Medicine, National Reference Centre for Rare Lymphatic and Vascular Diseases, UA11 INSERM - UM IDESP. Campus Santé, Montpellier Cedex 5, France
| |
Collapse
|
36
|
Okun SA, Lu D, Sew K, Subramaniam A, Lockwood WW. MET Activation in Lung Cancer and Response to Targeted Therapies. Cancers (Basel) 2025; 17:281. [PMID: 39858062 PMCID: PMC11764361 DOI: 10.3390/cancers17020281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
The hepatocyte growth factor receptor (MET) is a receptor tyrosine kinase (RTK) that mediates the activity of a variety of downstream pathways upon its activation. These pathways regulate various physiological processes within the cell, including growth, survival, proliferation, and motility. Under normal physiological conditions, this allows MET to regulate various development and regenerative processes; however, mutations resulting in aberrant MET activity and the consequent dysregulation of downstream signaling can contribute to cellular pathophysiology. Mutations within MET have been identified in a variety of cancers and have been shown to mediate tumorigenesis by increasing RTK activity and downstream signaling. In lung cancer specifically, a number of patients have been identified as possessing MET alterations, commonly receptor amplification (METamp) or splice site mutations resulting in loss of exon 14 (METex14). Due to MET's role in mediating oncogenesis, it has become an attractive clinical target and has led to the development of various targeted therapies, including MET tyrosine kinase inhibitors (TKIs). Unfortunately, these TKIs have demonstrated limited clinical efficacy, as patients often present with either primary or acquired resistance to these therapies. Mechanisms of resistance vary but often occur through off-target or bypass mechanisms that render downstream signaling pathways insensitive to MET inhibition. This review provides an overview of the therapeutic landscape for MET-positive cancers and explores the various mechanisms that contribute to therapeutic resistance in these cases.
Collapse
Affiliation(s)
- Sarah Anna Okun
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (S.A.O.); (K.S.); (A.S.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Daniel Lu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Katherine Sew
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (S.A.O.); (K.S.); (A.S.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Asha Subramaniam
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (S.A.O.); (K.S.); (A.S.)
- Department of Pathology and Laboratory Science, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - William W. Lockwood
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (S.A.O.); (K.S.); (A.S.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Pathology and Laboratory Science, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
37
|
Ashadul Sk M, K H, Matada GSP, Pal R, B V M, Mounika S, E H, M P V, D A. Current developments in PI3K-based anticancer agents: Designing strategies, biological activity, selectivity, structure-activity correlation, and docking insight. Bioorg Chem 2025; 154:108011. [PMID: 39662340 DOI: 10.1016/j.bioorg.2024.108011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024]
Abstract
The phospatidylinositol-3 kinase (PI3K) pathway is a critical intracellular signalling mechanism that is changed or amplified in a variety of cancers, including breast, gastric, ovarian, colorectal, prostate, glioma, and endometrial. PI3K signalling is important for cancer cell survival, angiogenesis, and metastasis, making it a promising therapeutic target. The PI3K kinases in their different isoforms, namely α, β, δ, and γ, encode PIK3CA, PIK3CB, PIK3CD, and PIK3CG genes. Specific gene mutation or overexpression of the protein is responsible for the therapeutic failure of current therapeutics. There are several current and completed clinical trials using PI3K inhibitors (pan, isoform-specific, and dual PI3K/mTOR) to develop effective PI3K inhibitors capable of overcoming resistance to existing drugs. However, the bulk of these inhibitors have had their indications revoked or voluntarily withdrawn due to concerns about their harmful consequences. Several inhibitors containing medicinally privileged scaffolds like thiazole, triazine, benzimidazole, podophyllotoxin, pyridine, quinazoline, thieno-triazole, pyrimidine, triazole, benzofuran, imidazo-pyridazine, oxazole, coumarin, and azepine derivatives have been explored to target the PI3K pathway and/or a specific isoform in the current overview. This article reviews the structure, biological activities, and clinical status of PI3K inhibitors. It focuses on the development techniques, docking insight, and structure-activity connections of PI3K-based inhibitors. The findings provide useful insights and future approaches for the development of promising PI3K-based inhibitors.
Collapse
Affiliation(s)
- Md Ashadul Sk
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru 560107, Karnataka, India
| | - Hemalatha K
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru 560107, Karnataka, India.
| | - Gurubasavaraja Swamy Purawarga Matada
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru 560107, Karnataka, India
| | - Rohit Pal
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru 560107, Karnataka, India.
| | - Manjushree B V
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru 560107, Karnataka, India
| | - S Mounika
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru 560107, Karnataka, India
| | - Haripriya E
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru 560107, Karnataka, India
| | - Viji M P
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru 560107, Karnataka, India
| | - Anjan D
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru 560107, Karnataka, India
| |
Collapse
|
38
|
Zhang Z, Deng J, Sun W, Wang Z. Cerebral Cavernous Malformation: From Genetics to Pharmacotherapy. Brain Behav 2025; 15:e70223. [PMID: 39740786 DOI: 10.1002/brb3.70223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2025] Open
Abstract
INTRODUCTION Cerebral cavernous malformation (CCM) is a type of cerebrovascular abnormality in the central nervous system linked to both germline and somatic genetic mutations. Recent preclinical and clinical studies have shown that various drugs can effectively reduce the burden of CCM lesions. Despite significant progress, the mechanisms driving CCM remain incompletely understood, and to date, no drugs have been developed that can cure or prevent CCM. This review aims to explore the genetic mutations, molecular mechanisms, and pharmacological interventions related to CCM. METHODS Literatures on the genetic mechanisms and pharmacological treatments of CCM can be searched in PubMed and Web of Science. RESULTS Germline and somatic mutations mediate the onset and development of CCM through several molecular pathways. Medications such as statins, fasudil, rapamycin, and propranolol can alleviate CCM symptoms or hinder its progression by specifically modulating the corresponding targets. CONCLUSIONS Understanding the molecular mechanisms underlying CCM offers potential for targeted therapies. Further research into novel mutations and treatment strategies is essential for improving patient outcomes.
Collapse
Affiliation(s)
- Zhuangzhuang Zhang
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Jianwen Deng
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Weiping Sun
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Zhaoxia Wang
- Department of Neurology, Peking University First Hospital, Beijing, China
| |
Collapse
|
39
|
Messing S, Widmeyer SRT, Denson JP, Mehalko J, Wall VE, Drew M, Snead K, Hong M, Grose C, Esposito D, Gillette W. Improved production of class I phosphatidylinositol 4,5-bisphosphate 3-kinase. Protein Expr Purif 2025; 225:106582. [PMID: 39173964 PMCID: PMC11421577 DOI: 10.1016/j.pep.2024.106582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/05/2024] [Accepted: 08/18/2024] [Indexed: 08/24/2024]
Abstract
Phosphatidylinositol 4,5-bisphosphate 3-kinases (PI3K) are a family of kinases whose activity affects pathways needed for basic cell functions. As a result, PI3K is one of the most mutated genes in all human cancers and serves as an ideal therapeutic target for cancer treatment. Expanding on work done by other groups we improved protein yield to produce stable and pure protein using a variety of modifications including improved solubility tag, novel expression modalities, and optimized purification protocol and buffer. By these means, we achieved a 40-fold increase in yield for p110α/p85α and a 3-fold increase in p110α. We also used these protocols to produce comparable constructs of the β and δ isoforms of PI3K. Increased yield enhanced the efficiency of our downstream high throughput drug discovery efforts on the PIK3 family of kinases.
Collapse
Affiliation(s)
- Simon Messing
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA.
| | - Stephanie R T Widmeyer
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - John-Paul Denson
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Jennifer Mehalko
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Vanessa E Wall
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Matthew Drew
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Kelly Snead
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Min Hong
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Carissa Grose
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Dominic Esposito
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - William Gillette
- Protein Expression Laboratory, NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| |
Collapse
|
40
|
Sheng Z, Beck P, Gabby M, Habte-Mariam S, Mitkos K. Molecular Basis of Oncogenic PI3K Proteins. Cancers (Basel) 2024; 17:77. [PMID: 39796708 PMCID: PMC11720314 DOI: 10.3390/cancers17010077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
The dysregulation of phosphatidylinositol 3-kinase (PI3K) signaling plays a pivotal role in driving neoplastic transformation by promoting uncontrolled cell survival and proliferation. This oncogenic activity is primarily caused by mutations that are frequently found in PI3K genes and constitutively activate the PI3K signaling pathway. However, tumorigenesis can also arise from nonmutated PI3K proteins adopting unique active conformations, further complicating the understanding of PI3K-driven cancers. Recent structural studies have illuminated the functional divergence among highly homologous PI3K proteins, revealing how subtle structural alterations significantly impact their activity and contribute to tumorigenesis. In this review, we summarize current knowledge of Class I PI3K proteins and aim to unravel the complex mechanism underlying their oncogenic traits. These insights will not only enhance our understanding of PI3K-mediated oncogenesis but also pave the way for the design of novel PI3K-based therapies to combat cancers driven by this signaling pathway.
Collapse
Affiliation(s)
- Zhi Sheng
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
- Department of Neurosurgery, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
- Faculty of Health Science, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Patrick Beck
- Division of General Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Maegan Gabby
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
| | | | - Katherine Mitkos
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
| |
Collapse
|
41
|
Wang W, Cheng Y, Meng Q, Jia B, Yao D, Cheng Y. The additive interaction of healthy lifestyles and genetic susceptibility on colorectal cancer risk in prediabetes: a large population-based prospective cohort study. BMC Gastroenterol 2024; 24:462. [PMID: 39696080 DOI: 10.1186/s12876-024-03552-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024] Open
Abstract
OBJECTIVE We aimed to investigate the interrelationships among polygenic risk scores (PRS), healthy lifestyle factors (HLFs), and colorectal cancer (CRC) risk in individuals with prediabetes. To investigate whether adherence to HLFs influence CRC risk in those with elevated PRS within this specific population. METHODS Data from 22,408 prediabetes participants without CRC at baseline were analyzed from the UK Biobank. HLFs were graded using healthy lifestyle scores (HLSs) and classified as favorable, intermediate, or unfavorable, while the PRS for CRC was categorized as high, medium, or low. Cox proportional hazards models were used to calculate hazard ratios (HR) and 95% confidence intervals (CI) for CRC risk. RESULTS High PRS (HR: 2.36; 95% CI: 1.86-3.00) and medium PRS (HR: 1.42; 95% CI: 1.09-1.83) prediabetes were associated with increased CRC risk compared to those with low PRS. HLFs were linked to lower CRC risk in a dose-response manner, with never smoking (HR: 0.69; 95% CI: 0.57-0.84) and maintaining a healthy BMI (HR: 0.64; 95% CI: 0.49-0.82) associated with reduced CRC risk. Adherence to favorable HLFs may reduce the CRC risk in those with medium (HR: 0.51; 95% CI: 0.27-0.95) and high PRS (HR: 0.62; 95% CI: 0.39-0.99) over 15 years of follow-up. In participants with high PRS and unfavorable HLFs, the excess risk due to the additive interaction between PRS and HLFs was 1.41% (p < 0.01), especially for women (1.07%). CONCLUSIONS There is an additive interaction of PRS and HLFs on CRC risk in individuals with prediabetes. Adopting favorable HLFs should be integrated into the management of prediabetes individuals to reduce the risk of CRC.
Collapse
Affiliation(s)
- Wenchen Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yikang Cheng
- The First Clinical Institute, Zunyi Medical University, Zunyi, 563000, China
| | - Qingyu Meng
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Baoqing Jia
- School of Medicine, Nankai University, Tianjin, 300071, China.
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Dawei Yao
- Department of Epidemiology and Biostatistics, School of Public Health, Xi'an, Jiaotong University Health Science Center, Xi'an, China.
| | - Yiping Cheng
- Department of Endocrinology, Shandong Provincial Hospital, Shandong First Medical University, Jinan, 250021, China.
| |
Collapse
|
42
|
Wang Y, Liu L, Graff SL, Cheng L. Recent advancements in biomarkers and molecular diagnostics in hormonal receptor-positive breast cancer. Histopathology 2024. [PMID: 39687977 DOI: 10.1111/his.15395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Molecular applications have limited use in breast cancer compared to other cancer types. In recent years, with an improving appreciation of the molecular genetics of breast cancer and innovative novel targeted and immune-mediated therapeutics, opportunities have arisen for more biomarker analysis and molecular applications in the diagnosis and treatment of both locally advanced and metastatic breast cancers. In hormone receptor-positive, HER2-negative breast cancers, a growing number of revolutionized personalized therapies are in clinical use or on trials, such as CDK4/6 inhibitors and immune checkpoint inhibitors in adjuvant and neoadjuvant settings, and PIK3CA inhibitors in metastatic disease. In this review, we focus on biomarkers associated with those new therapeutic targets and molecular applications for genetic alterations associated with drug resistance or interaction from a pathology perspective for selecting and optimizing breast cancer treatment.
Collapse
Affiliation(s)
- Yihong Wang
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Liu Liu
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Stephanie L Graff
- Division of Medical Oncology, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| |
Collapse
|
43
|
Bissegger L, Constantin TA, Keles E, Raguž L, Barlow-Busch I, Orbegozo C, Schaefer T, Borlandelli V, Bohnacker T, Sriramaratnam R, Schäfer A, Gstaiger M, Burke JE, Borsari C, Wymann MP. Rapid, potent, and persistent covalent chemical probes to deconvolute PI3Kα signaling. Chem Sci 2024; 15:20274-20291. [PMID: 39568927 PMCID: PMC11575505 DOI: 10.1039/d4sc05459h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/10/2024] [Indexed: 11/22/2024] Open
Abstract
Chemical probes have gained importance in the elucidation of signal transduction in biology. Insufficient selectivity and potency, lack of cellular activity and inappropriate use of chemical probes has major consequences on interpretation of biological results. The catalytic subunit of phosphoinositide 3-kinase α (PI3Kα) is one of the most frequently mutated genes in cancer, but fast-acting, high-quality probes to define PI3Kα's specific function to clearly separate it from other class I PI3K isoforms, are not available. Here, we present a series of novel covalent PI3Kα-targeting probes with optimized intracellular target access and kinetic parameters. On-target TR-FRET and off-target assays provided relevant kinetic parameters (k chem, k inact and K i) to validate our chemical probes. Additional intracellular nanoBRET tracer displacement measurements showed rapid diffusion across the cell membrane and extremely fast target engagement, while investigations of signaling downstream of PI3Kα via protein kinase B (PKB/Akt) and forkhead box O (FOXO) revealed blunted pathway activity in cancer cell lines with constitutively activated PI3Kα lasting for several days. In contrast, persistent PI3Kα inhibition was rapidly bypassed by other class I PI3K isoforms in cells lacking functional phosphatase and tensin homolog (PTEN). Comparing the rapidly-diffusing, fast target-engaging chemical probe 9 to clinical reversible PI3Kα-selective inhibitors alpelisib, inavolisib and 9r, a reversible analogue of 9, revealed 9's superior potency to inhibit growth (up to 600-fold) associated with sustained suppression of PI3Kα signaling in breast cancer cell lines. Finally, using a simple washout protocol, the utility of the highly-selective covalent PI3Kα probe 9 was demonstrated by the quantification of the coupling of insulin, EGF and CXCL12 receptors to distinct PI3K isoforms for signal transduction in response to ligand-dependent activation. Collectively, these findings along with the novel covalent chemical probes against PI3Kα provide insights into isoform-specific functions in cancer cells and highlight opportunities to achieve improved selectivity and long-lasting efficacy.
Collapse
Affiliation(s)
- Lukas Bissegger
- Department of Biomedicine, University of Basel Mattenstrasse 28 4058 Basel Switzerland +41 61 207 5046
| | - Theodora A Constantin
- Department of Biomedicine, University of Basel Mattenstrasse 28 4058 Basel Switzerland +41 61 207 5046
| | - Erhan Keles
- Department of Biomedicine, University of Basel Mattenstrasse 28 4058 Basel Switzerland +41 61 207 5046
| | - Luka Raguž
- Department of Biomedicine, University of Basel Mattenstrasse 28 4058 Basel Switzerland +41 61 207 5046
| | - Isobel Barlow-Busch
- Department of Biochemistry and Microbiology, University of Victoria Victoria British Columbia V8W 2Y2 Canada
| | - Clara Orbegozo
- Department of Biomedicine, University of Basel Mattenstrasse 28 4058 Basel Switzerland +41 61 207 5046
| | - Thorsten Schaefer
- Department of Biomedicine, University of Basel Mattenstrasse 28 4058 Basel Switzerland +41 61 207 5046
| | - Valentina Borlandelli
- Department of Biomedicine, University of Basel Mattenstrasse 28 4058 Basel Switzerland +41 61 207 5046
| | - Thomas Bohnacker
- Department of Biomedicine, University of Basel Mattenstrasse 28 4058 Basel Switzerland +41 61 207 5046
| | - Rohitha Sriramaratnam
- Department of Biomedicine, University of Basel Mattenstrasse 28 4058 Basel Switzerland +41 61 207 5046
| | - Alexander Schäfer
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich Otto-Stern-Weg 3 8093 Zürich Switzerland
| | - Matthias Gstaiger
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich Otto-Stern-Weg 3 8093 Zürich Switzerland
| | - John E Burke
- Department of Biochemistry and Microbiology, University of Victoria Victoria British Columbia V8W 2Y2 Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia Vancouver British Columbia V6T 1Z3 Canada
| | - Chiara Borsari
- Department of Biomedicine, University of Basel Mattenstrasse 28 4058 Basel Switzerland +41 61 207 5046
| | - Matthias P Wymann
- Department of Biomedicine, University of Basel Mattenstrasse 28 4058 Basel Switzerland +41 61 207 5046
| |
Collapse
|
44
|
Cokpinar S, Erdogdu IH, Orenay-Boyacioglu S, Boyacioglu O, Kahraman-Cetin N, Meteoglu I. PIK3CA Mutations and Co-Mutations in Operated Non-Small Cell Lung Carcinoma. J Clin Med 2024; 13:7472. [PMID: 39685930 DOI: 10.3390/jcm13237472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/16/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Understanding PIK3CA mutations and co-mutations in non-small cell lung carcinoma (NSCLC) is critical to developing personalized treatment strategies. Therefore, this study aims to investigate PIK3CA mutations and the accompanying somatic variations in NSCLC. Methods: This retrospective study included 98 patients over 18 years of age who were diagnosed with NSCLC, operated on, and referred to the Molecular Pathology Laboratory between January 2019 and June 2024 for next-generation sequencing panel tests and ALK-ROS1 FISH analysis. Results: All patients were found to carry PIK3CA mutations. Among the 98 NSCLC patients analyzed, 16 (16.33%) were female and 82 (83.67%) were male. The average age of the patients was 64.53 ± 9.63 years, with an age range of 38-84 years, and the majority were 50 years or older. Of the cases, 51 presented the adenocarcinoma subtype, while the remaining 47 showed the squamous cell carcinoma subtype. A smoking history was present in 77 (78.57%) patients, while 21 (21.43%) had no smoking history. The most frequently detected pathogenic or likely pathogenic PIK3CA variations were c.1633G>A p.E545K (32.65%), c.1624G>A p.E542K (11.22%), c.3140A>G p.H1047R (11.22%), c.3140A>T p.H1047L (5.10%), c.1357G>C p.E453Q (4.08%), and c.3143A>G p.H1048R (2.04%). The top 10 mutations that most commonly accompanied PIK3CA variations were KRAS, NF1, TP53, EGFR, PTEN, BRAF, KIT, CDKN2A, SMARCA4, and ATM mutations, respectively. Conclusions:PIK3CA variations, along with other gene variations, may influence cancer progression and thus may play a crucial role in the determination of targeted treatment strategies.
Collapse
Affiliation(s)
- Salih Cokpinar
- Department of Thoracic Surgery, School of Medicine, Aydin Adnan Menderes University, Aydin 09010, Türkiye
| | - Ibrahim Halil Erdogdu
- Department of Molecular Pathology, School of Medicine, Aydin Adnan Menderes University, Aydin 09010, Türkiye
| | - Seda Orenay-Boyacioglu
- Department of Medical Genetics, School of Medicine, Aydin Adnan Menderes University, Aydin 09010, Türkiye
| | - Olcay Boyacioglu
- Faculty of Engineering, Aydin Adnan Menderes University, Aydin 09010, Türkiye
| | - Nesibe Kahraman-Cetin
- Department of Molecular Pathology, School of Medicine, Aydin Adnan Menderes University, Aydin 09010, Türkiye
| | - Ibrahim Meteoglu
- Department of Molecular Pathology, School of Medicine, Aydin Adnan Menderes University, Aydin 09010, Türkiye
| |
Collapse
|
45
|
Yasin F, Sokol E, Vasan N, Pavlick DC, Huang RSP, Pelletier M, Levy MA, Pusztai L, Lacy J, Zhang JY, Ross JS, Cecchini M. Molecular characteristics of advanced colorectal cancer and multi-hit PIK3CA mutations. Oncologist 2024; 29:1059-1067. [PMID: 39401325 PMCID: PMC11630746 DOI: 10.1093/oncolo/oyae259] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 08/23/2024] [Indexed: 12/12/2024] Open
Abstract
INTRODUCTION Approximately 20% of patients living with colorectal cancer (CRC) have activating mutations in their tumors in the PIK3CA oncogene. Two or more activating mutations (multi-hit) for the PIK3CA allele increase PI3K⍺ signaling compared to single-point mutations, resulting in exceptional response to PI3K⍺ inhibition. We aimed to identify the prevalence of PIK3CA multi-hit mutations in metastatic CRC to identify patients who may benefit from PI3K inhibitors. METHODS The Foundation Medicine database (Boston, MA, USA) was analyzed for patients with CRC who underwent genomic profiling on tumor DNA isolated during routine clinical care from 2013 to 2021. Molecular and clinical variables were abstracted for patients with PIK3CA mutations. RESULTS We identified 49 051 patients with CRC who underwent Foundation Medicine testing. 710/41154 (1.7%) patients had multi-hit PIK3CA mutations, of which 53% were male (n = 448) with a median age of 60. Microsatellite status was available for 697 patients with multi-hit PIK3CA and 17.6% (123/697) were microsatellite instability-high. Clinically relevant mutations in KRAS and BRAFV600E were seen in 459/710 (64.7%) and 65/710 (9.1%), respectively. The 4 most common PIK3CA variants were H1047R (9.8%), E545K (9.2%), E542K (9.0%), and R88Q (7.1%). The most common variant pair was E542K-E545K (4.7%). CONCLUSIONS Multi-hit mutations in PIK3CA are seen in 1.7% of advanced CRC, a meaningful prevalence given the high burden of CRC worldwide, and may represent a subset of patients that have enhanced sensitivity to PI3K inhibition. Future investigation regarding the clinical utility of PI3K inhibitors is warranted in multi-hit PIK3CA CRC.
Collapse
Affiliation(s)
- Faiza Yasin
- Department of Medicine (Medical Oncology), Yale University, New Haven, CT 06510, United States
- Yale Cancer Center, New Haven, CT 06510, United States
| | - Ethan Sokol
- Foundation Medicine, Boston, MA 02210, United States
| | - Neil Vasan
- Department of Medicine (Hematology/Oncology), Columbia University, New York, NY 10032, United States
| | | | | | | | | | - Lajos Pusztai
- Department of Medicine (Medical Oncology), Yale University, New Haven, CT 06510, United States
- Yale Cancer Center, New Haven, CT 06510, United States
| | - Jill Lacy
- Department of Medicine (Medical Oncology), Yale University, New Haven, CT 06510, United States
- Yale Cancer Center, New Haven, CT 06510, United States
| | - Janie Yue Zhang
- Department of Medicine (Medical Oncology), Yale University, New Haven, CT 06510, United States
- Department of Medicine (Medical Oncology), University of Pittsburgh Medical Center, Pittsburgh, PA 15219, United States
| | - Jeffrey S Ross
- Foundation Medicine, Boston, MA 02210, United States
- Upstate Medical University, Syracuse, NY 13210, United States
| | - Michael Cecchini
- Department of Medicine (Medical Oncology), Yale University, New Haven, CT 06510, United States
- Yale Cancer Center, New Haven, CT 06510, United States
| |
Collapse
|
46
|
Martínez-Rodríguez A, Fuentes-Antrás J, Lorca V, López de Sá A, Pérez-Segura P, Moreno F, García-Sáenz JA, García-Barberán V. Molecular Profiling of Endocrine Resistance in HR+/HER2-Metastatic Breast Cancer: Insights from Extracellular Vesicles-Derived DNA and ctDNA in Liquid Biopsies. Int J Mol Sci 2024; 25:13045. [PMID: 39684756 DOI: 10.3390/ijms252313045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/27/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024] Open
Abstract
Standard treatments in hormone receptor-positive (HR+)/HER2-metastatic breast cancer (mBC) typically involve endocrine therapy (ET) combined with CDK4/6 inhibitors, yet resistance to ET remains a persistent challenge in advanced cases. A deeper knowledge of the use of liquid biopsy is crucial for the implementation of precision medicine in mBC with real-time treatment guidance. Our study assesses the prognostic value of PIK3CA and ESR1 mutations in DNA derived from extracellular vesicles (EV-DNA) in longitudinal plasma from 59 HR+/HER2-mBC patients previously exposed to aromatase inhibitors, with a comparative analysis against circulating tumor DNA (ctDNA). Mutations were evaluated by digital PCR. PIK3CA and ESR1 mutations were found in 22 and 25% of patients. Baseline ESR1 mutations in EV-DNA were associated with shorter progression-free survival (PFS) across the cohort, with the Y537S mutation showing a particularly strong impact on the outcome of fulvestrant-treated patients. In contrast, PIK3CA mutations in EV-DNA did not significantly correlate with PFS, whereas in ctDNA, they were linked to poor outcomes. Altogether, this study positions EV-DNA as a valuable biomarker alongside ctDNA, enriching the understanding of different analytes in liquid biopsy and supporting strategies for HR+/HER2-mBC in precision oncology.
Collapse
MESH Headings
- Humans
- Female
- Breast Neoplasms/genetics
- Breast Neoplasms/drug therapy
- Breast Neoplasms/pathology
- Breast Neoplasms/blood
- Extracellular Vesicles/metabolism
- Extracellular Vesicles/genetics
- Circulating Tumor DNA/genetics
- Circulating Tumor DNA/blood
- Liquid Biopsy/methods
- Middle Aged
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Class I Phosphatidylinositol 3-Kinases/genetics
- Drug Resistance, Neoplasm/genetics
- Aged
- Mutation
- Estrogen Receptor alpha/genetics
- Estrogen Receptor alpha/metabolism
- Biomarkers, Tumor/genetics
- Adult
- Aromatase Inhibitors/therapeutic use
- Aromatase Inhibitors/pharmacology
- Prognosis
- Receptors, Estrogen/metabolism
- Receptors, Estrogen/genetics
- Receptors, Progesterone/metabolism
- Receptors, Progesterone/genetics
- Neoplasm Metastasis
- Antineoplastic Agents, Hormonal/therapeutic use
- Antineoplastic Agents, Hormonal/pharmacology
Collapse
Affiliation(s)
- Ana Martínez-Rodríguez
- "Clinical and Translational Research in Oncology" Group, Molecular Oncology Laboratory, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria Hospital Clinico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Medical Oncology, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria Hospital Clinico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Jesús Fuentes-Antrás
- Department of Medical Oncology, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria Hospital Clinico San Carlos (IdISSC), 28040 Madrid, Spain
- NEXT Oncology Experimental Therapeutics Unit, Hospital Universitario Quironsalud Madrid, 28223 Madrid, Spain
| | - Víctor Lorca
- "Clinical and Translational Research in Oncology" Group, Molecular Oncology Laboratory, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria Hospital Clinico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Alfonso López de Sá
- Department of Medical Oncology, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria Hospital Clinico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Pedro Pérez-Segura
- Department of Medical Oncology, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria Hospital Clinico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Fernando Moreno
- Department of Medical Oncology, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria Hospital Clinico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Jose Angel García-Sáenz
- Department of Medical Oncology, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria Hospital Clinico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Vanesa García-Barberán
- "Clinical and Translational Research in Oncology" Group, Molecular Oncology Laboratory, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria Hospital Clinico San Carlos (IdISSC), 28040 Madrid, Spain
| |
Collapse
|
47
|
Zhu S, Yu D, Wang X, Wang X. Predict the Drug-Drug Interaction of a Novel PI3Kα/δ Inhibitor, TQ-B3525, and Its Two Metabolites Using Physiologically Based Pharmacokinetic Modeling. J Clin Pharmacol 2024; 64:1517-1527. [PMID: 39105511 DOI: 10.1002/jcph.6111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/22/2024] [Indexed: 08/07/2024]
Abstract
A novel dual PI3K α/δ inhibitor, TQ-B3525, has been developed for the targeted treatment of lymphoma and solid tumors. TQ-B3525 is primarily metabolized by CYP3A4 and FOM3, while also serving as a substrate for the P-glycoprotein transporter. The aim of this study was to anticipate the drug-drug interaction (DDI) of TQ-B3525 and its two metabolites with CYP3A4 enzyme potent inducer (rifampicin) and CYP3A4/P-gp inhibitor (itraconazole) utilizing a physiologically based pharmacokinetic (PBPK) modeling approach. Clinical data from healthy and cancer patient adults were employed to construct and evaluate the PBPK model for TQ-B3525, M3, and M8-3. Models involving rifampicin combined with midazolam, itraconazole combined with midazolam or digoxin were utilized to showcase the robustness of evaluating DDI effects. The simulated drug exposure of TQ-B3525, M3, and M8-3 in healthy and patient adults were consistent with clinical data, and the mean fold error values were within the acceptable ranges. The simulated results of positive substrates correspond to those reported in the literature. Co-administration with rifampicin reduces Cmax and AUC of TQ-B3525 to 76.1% and 46.0%, while increasing the levels of M3 and M8-3. With itraconazole, Cmax and AUC of TQ-B3525 rise to 131% and 204%, but decrease substantially for M3 and M8-3. PBPK model simulation results showed that the systemic exposure of TQ-B3525 was significantly affected when co-administered with CYP3A4/P-gp inducers and inhibitors. This indicates that the combination with strong inducers and inhibitors should be carefully avoided or adjust the dosage of TQ-B3525 in clinic.
Collapse
Affiliation(s)
- Shixing Zhu
- Clinical Medicine Department, Chia Tai Tianqing Pharmaceutical Group Co., Ltd., Nanjing, China
| | - Ding Yu
- Clinical Medicine Department, Chia Tai Tianqing Pharmaceutical Group Co., Ltd., Nanjing, China
| | - Xunqiang Wang
- Clinical Medicine Department, Chia Tai Tianqing Pharmaceutical Group Co., Ltd., Nanjing, China
| | - Xin Wang
- Clinical Medicine Department, Chia Tai Tianqing Pharmaceutical Group Co., Ltd., Nanjing, China
| |
Collapse
|
48
|
Astore S, Oneda E, Zaniboni A. A therapeutic algorithm guiding subsequent therapy selection after CDK4/6 inhibitors' failure: A review of current and investigational treatment for HR+/Her2- breast cancer. Crit Rev Oncol Hematol 2024; 204:104535. [PMID: 39433229 DOI: 10.1016/j.critrevonc.2024.104535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/01/2024] [Accepted: 10/06/2024] [Indexed: 10/23/2024] Open
Abstract
The first-line combination therapies utilizing cyclin-dependent kinase 4 and 6 inhibitors (CDK4/6i) with endocrine therapy (ET) have significantly impacted the course of hormone receptor positive (HR+)/Human Epidermal Growth Factor Receptor 2 negative (HER2-) advanced breast cancer (ABC). However, resistance often emerges, leading to a molecularly different disease. Estrogen receptor one (ESR1) gene mutations, driving resistance to aromatase inhibitors (AIs), may guide the use of fulvestrant or emerging oral selective estrogen receptor degraders (SERDs) like elacestrant. The dynamic nature of ESR1 mutations suggests potential guidance for continuing CDK4/6i therapy beyond progression. Targeting mutations like breast cancer gene 1 and 2 (BRCA 1/2) with Poly (ADP-ribose) polymerase (PARP) inhibitors or the PI3K/AKT/mTOR pathway provides therapeutic options. The advent of antibody-drug conjugates (ADCs) like trastuzumab deruxtecan (T-DXd) and novel agents targeting Trophoblast cell surface antigen-2 (Trop-2) introduces further complexity, underscoring the need for early intervention targeting specific genomic alterations in metastatic BC.
Collapse
Affiliation(s)
- Serena Astore
- Medical Oncology Dept. Fondazione Poliambulanza, Brescia, Italy.
| | - Ester Oneda
- Medical Oncology Dept. Fondazione Poliambulanza, Brescia, Italy
| | | |
Collapse
|
49
|
Utpal BK, Dehbia Z, Zidan BMRM, Sweilam SH, Singh LP, Arunkumar MS, Sona M, Panigrahy UP, Keerthana R, Mandadi SR, Rab SO, Alshehri MA, Koula D, Suliman M, Nafady MH, Emran TB. Carotenoids as modulators of the PI3K/Akt/mTOR pathway: innovative strategies in cancer therapy. Med Oncol 2024; 42:4. [PMID: 39549201 DOI: 10.1007/s12032-024-02551-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 10/29/2024] [Indexed: 11/18/2024]
Abstract
Cancer progression is primarily driven by the uncontrolled activation of cellular signaling pathways, with the PI3K/Akt/mTOR (PAMT) pathway playing a central role. This pathway significantly contributes to the proliferation and survival of cancer cells, and its hyperactivity is a major challenge in managing several types of malignancies. This article delves into the promising potential of carotenoids, natural pigments found in abundance in fruits and vegetables, as a novel therapeutic strategy for cancer treatment. By specifically targeting and inhibiting the PAMT pathway, carotenoids may effectively disrupt the growth and survival of cancer cells. The article examines the complex mechanisms underlying these interactions and highlights the obstacles faced in cancer treatment. It proposes a compelling approach to developing therapies that leverage natural products to target this critical pathway, offering a fresh perspective on cancer treatment. Further research is essential to enhance the therapeutic efficacy of these compounds.
Collapse
Affiliation(s)
- Biswajit Kumar Utpal
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Zerrouki Dehbia
- Laboratory of AgroBiotechnology and Nutrition in Semi Arid Zones, Faculty of Nature and Life Sciences, University of Ibn Khaldoun, Tiaret, Algeria
| | - B M Redwan Matin Zidan
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, Cairo, 11829, Egypt
| | - Laliteshwar Pratap Singh
- Department of Pharmaceutical Chemistry, Narayan Institute of Pharmacy, Gopal Narayan Singh University, Sasaram (Rohtas) Bihar, Jamuhar, 821305, India
| | - M S Arunkumar
- Faculty of Pharmacy, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, 641021, India
| | - M Sona
- Faculty of Pharmacy, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, 641021, India
| | - Uttam Prasad Panigrahy
- Faculty of Pharmaceutical Science, Assam Down Town University, Gandhi Nagar, Sankar Madhab Path, Panikhaiti, Guwahati, Assam, India
| | - R Keerthana
- Faculty of Pharmacy, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, 641021, India
| | - Sandhya Rani Mandadi
- Department of Pharmaceutics, Vishnu Institute of Pharmaceutical Education and Research, Tuljaraopet, Telangana , 502313, India
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Mohammed Ali Alshehri
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Doukani Koula
- Laboratory of AgroBiotechnology and Nutrition in Semi Arid Zones, Faculty of Nature and Life Sciences, University of Ibn Khaldoun, Tiaret, Algeria
- Laboratory of Animal Production Sciences and Techniques, University of Abdelhamid Ibn Badis, Mostaganem, Algeria
| | - Muath Suliman
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Mohamed H Nafady
- Faculty of Applied Health Science Technology, Misr University for Science and Technology, Giza, 12568, Egypt.
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| |
Collapse
|
50
|
Li SL, Wang PY, Jia YP, Zhang ZX, He HY, Chen PY, Liu X, Liu B, Lu L, Fu WH. BIRC3 induces the phosphoinositide 3-kinase-Akt pathway activation to promote trastuzumab resistance in human epidermal growth factor receptor 2-positive gastric cancer. World J Gastrointest Oncol 2024; 16:4436-4455. [PMID: 39554734 PMCID: PMC11551635 DOI: 10.4251/wjgo.v16.i11.4436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/13/2024] [Accepted: 09/10/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Trastuzumab-targeted therapy is currently the standard of care for advanced human epidermal growth factor receptor 2 (HER2)-positive gastric cancer. However, the emergence of resistance to trastuzumab poses significant challenges. AIM To identify the key genes associated with trastuzumab resistance. These results provide a basis for the development of interventions to address drug resistance and improve patient outcomes. METHODS High-throughput sequencing and bioinformatics were used to identify the differentially expressed pivotal gene BIRC3 and delineate its potential function and pathway regulation. Tumor samples were collected from patients with HER2-positive gastric cancer to evaluate the correlation between BIRC3 expression and trastuzumab resistance. We established gastric cancer cell lines with both highly expressed and suppressed levels of BIRC3, followed by comprehensive in vitro and in vivo experiments to confirm the involvement of BIRC3 in trastuzumab resistance and to elucidate its underlying mechanisms. RESULTS In patients with HER2-positive gastric cancer, there is a significant correlation between elevated BIRC3 expression in tumor tissues and higher T stage, tumor node metastasis stage, as well as poor overall survival and progression-free survival. BIRC3 is highly expressed in trastuzumab-resistant gastric cancer cell lines, where it inhibits tumor cell apoptosis and enhances trastuzumab resistance by promoting the phosphorylation and activation of the phosphoinositide 3-kinase-Akt (PI3K-AKT) pathway in HER2-positive gastric cancer cells, both in vivo and in vitro. CONCLUSION This study revealed a robust association between high BIRC3 expression and an unfavorable prognosis in patients with HER2-positive gastric cancer. Thus, the high expression of BIRC3 stimulated PI3K-AKT phosphorylation and activation, stimulating the proliferation of HER2-positive tumor cells and suppressing apoptosis, ultimately leading to trastuzumab resistance.
Collapse
Affiliation(s)
- Shu-Liang Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300052, China
- Department of Gastrointestinal Surgery, The Second People’s Hospital of Liaocheng, Liaocheng 252600, Shandong Province, China
- Department of Gastrointestinal Surgery, The Second Hospital of Liaocheng, Affiliated to Shandong First Medical University, Liaocheng 252600, Shandong Province, China
| | - Pei-Yao Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Yang-Pu Jia
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Zhao-Xiong Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Hao-Yu He
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Peng-Yu Chen
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Xin Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Bang Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Li Lu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Wei-Hua Fu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300052, China
| |
Collapse
|