Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,118)

Search Parameters:
Keywords = PARP inhibitor

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
13 pages, 948 KB  
Article
The Real-World Impact of PARP Inhibitor Maintenance Therapy in High Grade Serous Tubo-Ovarian and Peritoneal Cancers
by Maryam Al-Ani, Bahaaeldin Baraka, Navin Mathiyalagan, Muhammad Adeel Sarwar, Avinash Segaran, Wafaa Abuzahra, Alayna Radford, Kersty Buxton, Lalith Seneviratne, Santhanam Sundar, Anjana Anand, David Nunns, Karin Williamson, Ben Wormald, Ketankumar Gajjar and Srinivasan Madhusudan
Cancers 2025, 17(21), 3591; https://doi.org/10.3390/cancers17213591 - 6 Nov 2025
Viewed by 460
Abstract
Background: Pivotal clinical trials have led to the routine clinical use of PARP inhibitor (PARPi) (olaparib, niraparib, or rucaparib) maintenance therapy in high-grade serous tubo-ovarian and peritoneal cancers. Whether various PARPis have comparable clinical impact in the real-world is an area of ongoing [...] Read more.
Background: Pivotal clinical trials have led to the routine clinical use of PARP inhibitor (PARPi) (olaparib, niraparib, or rucaparib) maintenance therapy in high-grade serous tubo-ovarian and peritoneal cancers. Whether various PARPis have comparable clinical impact in the real-world is an area of ongoing investigation. Methods: We conducted a retrospective study of all patients who received PARPi maintenance therapy at Nottingham Cancer Centre from October 2017 to April 2025. Clinical data were extracted from multidisciplinary team electronic health records, including age, BRCA mutation status, HRD status, treatment history, type of PARP inhibitor received, progression-free survival (PFS), and overall survival (OS). Results: A total of 177 patients had received PARPi therapy with a mean age of 63 years at diagnosis. In all, 94/177 (53.1%) had received PARPi as primary maintenance, while 83/177 (46.9%) were treated in the recurrent setting. All together, 25/177 (14.1%) had BRCA1 germline mutation and 21/177 (11.9%) had BRCA2 germline mutation. In the primary olaparib setting, PFS was significantly better in BRCA2 germline-mutated patients compared to BRCA1 germline-mutated patients [median PFS was not reached vs. 29.0 months, respectively, p = 0.002]. In BRCA, wild-type patients receiving primary niraparib, median PFS was 11 months. Median PFS for patients with upfront surgery was 37 months compared to 19 months in the interval debulking group but not significant (p = 0.49). In the recurrent setting, there was no significant difference in median PFS between niraparib and rucaparib [10 months vs. 9 months, p = 0.594]. Conclusions: BRCA2 germline-mutated patients obtained significantly greater benefit from olaparib compared to BRCA1-mutated patients. PFS benefit from niraparib (primary or recurrent setting) is comparable to clinical trials. There was no difference in benefit between niraparib and rucaparib in the recurrent setting. Full article
(This article belongs to the Special Issue Advances in Ovarian Cancer Treatment: Past, Present and Future)
Show Figures

Figure 1

15 pages, 1506 KB  
Review
Computational Chemistry Advances in the Development of PARP1 Inhibitors for Breast Cancer Therapy
by Charmy Twala, Penny Govender and Krishna Govender
Pharmaceuticals 2025, 18(11), 1679; https://doi.org/10.3390/ph18111679 - 6 Nov 2025
Viewed by 465
Abstract
Poly (ADP-ribose) polymerase 1 (PARP1) is an important enzyme that plays a central role in the DNA damage response, facilitating repair of single-stranded DNA breaks via the base excision repair (BER) pathway and thus genomic integrity. Its therapeutic relevance is compounded in breast [...] Read more.
Poly (ADP-ribose) polymerase 1 (PARP1) is an important enzyme that plays a central role in the DNA damage response, facilitating repair of single-stranded DNA breaks via the base excision repair (BER) pathway and thus genomic integrity. Its therapeutic relevance is compounded in breast cancer, particularly in BRCA1 or BRCA2 mutant cancers, where compromised homologous recombination repair (HRR) leaves a synthetic lethal dependency on PARP1-mediated repair. This review comprehensively discusses the recent advances in computational chemistry for the discovery of PARP1 inhibitors, focusing on their application in breast cancer therapy. Techniques such as molecular docking, molecular dynamics (MD) simulations, quantitative structure–activity relationship (QSAR) modeling, density functional theory (DFT), time-dependent DFT (TD-DFT), and machine learning (ML)-aided virtual screening have revolutionized the discovery of inhibitors. Some of the most prominent examples are Olaparib (IC50 = 5 nM), Rucaparib (IC50 = 7 nM), and Talazoparib (IC50 = 1 nM), which were optimized with docking scores between −9.0 to −9.3 kcal/mol and validated by in vitro and in vivo assays, achieving 60–80% inhibition of tumor growth in BRCA-mutated models and achieving up to 21-month improvement in progression-free survival in clinical trials of BRCA-mutated breast and ovarian cancer patients. These strategies enable site-specific hopping into the PARP1 nicotinamide-binding pocket to enhance inhibitor affinity and specificity and reduce off-target activity. Employing computation and experimental verification in a hybrid strategy have brought next-generation inhibitors to the clinic with accelerated development, higher efficacy, and personalized treatment for breast cancer patients. Future approaches, including AI-aided generative models and multi-omics integration, have the promise to further refine inhibitor design, paving the way for precision oncology. Full article
Show Figures

Graphical abstract

14 pages, 258 KB  
Review
Breast-Conserving Surgery in BRCA Mutation Carriers: A Paradigm Shift Toward Individualized, Multidisciplinary Care
by Calogero Cipolla, Giuseppa Scandurra, Daniela Sambataro, Chiara Mesi, Martina Greco, Eleonora D’Agati, Vittorio Gebbia, Luca Giacomelli and Maria Rosaria Valerio
Life 2025, 15(11), 1701; https://doi.org/10.3390/life15111701 - 3 Nov 2025
Viewed by 404
Abstract
Breast cancer associated with BRCA1 and BRCA2 mutations presents unique therapeutic challenges, traditionally favoring mastectomy due to concerns over recurrence and new primaries. However, evolving evidence and advances in multimodal therapy have reshaped this paradigm, positioning breast-conserving surgery (BCS) as a viable option [...] Read more.
Breast cancer associated with BRCA1 and BRCA2 mutations presents unique therapeutic challenges, traditionally favoring mastectomy due to concerns over recurrence and new primaries. However, evolving evidence and advances in multimodal therapy have reshaped this paradigm, positioning breast-conserving surgery (BCS) as a viable option for selected carriers. This narrative review synthesizes current data from meta-analyses, retrospective cohorts, and pivotal studies, including a multicenter analysis which affirmed oncologic equivalence between BCS and mastectomy when combined with radiotherapy and systemic therapy. While meta-analyses confirm higher local events following BCS, survival remains comparable, indicating that recurrence reflects genetic predisposition rather than surgical inadequacy. Optimized systemic treatments, including chemotherapy, endocrine therapy, risk-reducing salpingo-oophorectomy, and PARP inhibitors, further mitigate recurrence risk. Meanwhile, patient-centered outcomes favor BCS: studies consistently link it to improved body image, psychosocial well-being, and quality of life, especially for younger BRCA carriers. Fertility-preserving options remain viable, with evidence supporting the safety of pregnancy, breastfeeding, and assisted reproductive technologies in BRCA-mutated survivors. These findings support individualized surgical planning for BRCA carriers within multidisciplinary care, balancing oncologic safety, systemic strategies, and psychosocial priorities. BCS should be considered a standard option for well-selected patients in hereditary breast cancer management. Full article
15 pages, 4825 KB  
Article
Radiosensitization Effect of PARP Inhibitor Talazoparib Involves Decreasing Mitochondrial Membrane Potential and Induction of Cellular Senescence
by Barkha Saraswat, Ankitha Vadi Velu, Zhongming Gao, Zongxiang Zhang, Haoyang Zhu, Ying Tong and Mitsuko Masutani
Curr. Issues Mol. Biol. 2025, 47(11), 908; https://doi.org/10.3390/cimb47110908 - 1 Nov 2025
Viewed by 342
Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors (PARPis) with radiation therapy can enhance the sensitivity of cancer cells by inhibiting DNA repair pathways. To determine the most suitable PARP inhibitor for radiosensitization in cancer cells, we compared various types of clinically used PARPis in lung [...] Read more.
Poly (ADP-ribose) polymerase (PARP) inhibitors (PARPis) with radiation therapy can enhance the sensitivity of cancer cells by inhibiting DNA repair pathways. To determine the most suitable PARP inhibitor for radiosensitization in cancer cells, we compared various types of clinically used PARPis in lung cancer A549 cells. We found that most PARP inhibitors showed radiosensitization effects on A549 cells. ER10 values for talazoparib, olaparib rucaparib, ABT888 and niraparib were 1.5, 1.8, 2.8, 1.4, and 1.4, respectively. Talazoparib showed a radiosensitization effect at its lowest concentration. Talazoparib is a potent PARP inhibitor and has been used in clinical settings for several types of cancer as an anti-cancer agent. We thus focused on how talazoparib causes radiosensitization in lung cancer A549 cells. As a result of the combination of talazoparib and γ-irradiation, we observed an increased level of cellular senescence accompanied by a decrease in mitochondrial membrane potential. When the p21 gene was knocked down, both the decrease in mitochondrial membrane potential and senescence level were attenuated, suggesting that p21 is involved in senescence induction after γ-irradiation combined with talazoparib treatment. Taken together, we showed that PARP inhibitor talazoparib treatment in combination with γ-irradiation causes cellular senescence in lung cancer cells, involving p21 function. Full article
(This article belongs to the Special Issue Radiation-Induced Cellular and Molecular Responses)
Show Figures

Figure 1

20 pages, 5991 KB  
Article
Combinative Treatment of the PARP Inhibitor Olaparib and Antimetastasis Ruthenium(II)–Arene Compound RAPTA-T for Triple-Negative BRCA1 Wild-Type Breast Cancer Cells
by Adisorn Ratanaphan
Int. J. Mol. Sci. 2025, 26(21), 10613; https://doi.org/10.3390/ijms262110613 - 31 Oct 2025
Viewed by 346
Abstract
To date, breast cancer remains one of the leading causes of death among women worldwide. Although various treatments are used in clinical settings, the efficacy and safety of such treatments are limited by tumor biology factors and patient preferences. Previous studies have shown [...] Read more.
To date, breast cancer remains one of the leading causes of death among women worldwide. Although various treatments are used in clinical settings, the efficacy and safety of such treatments are limited by tumor biology factors and patient preferences. Previous studies have shown that triple-negative BRCA1-deficient breast cancer is susceptible to DNA-damaging agents, including platinum-based drugs and poly(ADP-ribose) polymerase (PARP) inhibitors, alone or in combination. To address whether the combinative treatment of these DNA-damaging agents can be extended to the triple-negative BRCA1-proficient breast cancer population, we investigated the anticancer activity of the well-known FDA-approved PARP inhibitor olaparib in combination with the antimetastatic ruthenium(II)–arene PTA compound RAPTA-T for triple-negative BRCA1-competent breast cancer cells (MDA-MB-468 and MDA-MB-231), with consideration of sporadic breast cancer MCF-7 cells. RAPTA-T, olaparib, and the combined agents exhibited a dose-dependent inhibition of breast cancer cell growth in selected breast cancer cells. The combination compound inhibited colony formation most effectively in MDA-MB-468 cells. Additionally, the scratch-wound assay showed that MDA-MB-468 cells migrated more slowly than MCF-7 and MDA-MB-231 cells. The results indicated that the olaparib and RAPTA-T combination can reduce or inhibit the survival, invasion, and metastasis of breast cancer cells. Moreover, the combined agents promoted apoptotic cell death, with a higher percentage of apoptosis observed in MDA-MB-468 cells than in MDA-MB-231 and MCF-7 cells. Olaparib and RAPTA-T also interfered with cell cycle progression, with the greatest inhibition observed in the S and G2/M phases of MCF-7 cells (1.6- and 3.4-fold), followed by MDA-MB-468 cells (1.6- and 1.8-fold) and MDA-MB-231 cells (1.5- and 1.4-fold). Interestingly, MDA-MB-468 cells presented the highest degree of inhibition for BRCA1 replication and BRCA1 expression. The p53, PARP, and Chk1 proteins were more strongly upregulated in MDA-MB-231 cells than in Ru-untreated control cells. Moreover, the expression levels of protein biomarkers associated with the epithelial-to-mesenchymal transition (EMT), including E-cadherin and SLUG, were remarkably reduced in all tested breast cancer cells. Together, our results show the feasibility of extending the application of PARP inhibitors beyond breast cancer with BRCA1 mutations and optimizing the combinative treatment of PARP inhibitors with antimetastasis ruthenium-based chemotherapy as new therapeutic approaches for TNBC harboring wild-type BRCA1. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

20 pages, 6072 KB  
Article
Reversing the Warburg Effect: YW3-56 Induces Leukemia Differentiation via AKT-Mediated Glucose Metabolic Reprogramming
by Di Zhu, Dan Gao, Yu Lu, Na Chen, Li Zhang, Lan Zhang and Yuji Wang
Pharmaceuticals 2025, 18(11), 1646; https://doi.org/10.3390/ph18111646 - 31 Oct 2025
Viewed by 287
Abstract
Background: Protein arginine deiminase 4 (PAD4) has emerged as a promising therapeutic target for acute promyelocytic leukemia (APL) because of its role in epigenetic regulation and leukemogenesis. All-trans retinoic acid, a standard differentiation agent in APL therapy, has been shown to upregulate [...] Read more.
Background: Protein arginine deiminase 4 (PAD4) has emerged as a promising therapeutic target for acute promyelocytic leukemia (APL) because of its role in epigenetic regulation and leukemogenesis. All-trans retinoic acid, a standard differentiation agent in APL therapy, has been shown to upregulate PAD4 expression during leukemic cell maturation. Interestingly, first-generation PAD4 inhibitors also promote differentiation, but simultaneously trigger compensatory PAD4 overexpression, underscoring the unresolved complexity of PAD4 modulation in leukemia therapy. Methods: In this study, we employed mass cytometry and transcriptomic–proteomic integrated analysis to investigate the underlying mechanisms of YW3-56, a dual-function PAD4 inhibitor against protein expression and enzymatic function, in NB4 leukemia cells. Functional validation was conducted using Western blot and metabolic assays. Results: Mass cytometry analysis revealed that YW3-56 reduced leukemia stemness (CD44/CD133), while enhancing myeloid differentiation (CD11b/CD14) and immunogenic activation (CD80/CD86). Multiomics analysis revealed a YW3-56-induced metabolic shift characterized by downregulation of glycolytic enzymes and upregulation of the tricarboxylic acid cycle and pentose phosphate pathway components, indicating a reversal of the Warburg effect. Mechanistically, this metabolic reprogramming was driven by reduced AKT expression and phosphorylation at Thr308, impaired GLUT1 expression and membrane localization, and decreased glucose uptake, which collectively promoted the differentiation of NB4 cells. Additionally, YW3-56 suppressed the downstream mTOR pathway, inducing caspase-3/PARP-mediated apoptosis and inhibiting cell proliferation. Conclusions: Our study demonstrated that YW3-56 exerts multimodal antileukemic effects in APL by simultaneously targeting PAD4-mediated epigenetic regulation, AKT-driven metabolic reprogramming and cellular differentiation, highlighting PAD4-AKT signaling as a promising target for APL combination therapy. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Graphical abstract

39 pages, 12648 KB  
Article
The SP1-SuperEnhancer-SPHK1 Axis Mediates Niraparib Resistance in TNBC
by Yu-Xia Yuan, Rui-Jia Chen, Gui-Hui Tu, Chao-Qi Li, Long-Long Xu, Yi-Ling Lu and Li-Xian Wu
Pharmaceuticals 2025, 18(11), 1622; https://doi.org/10.3390/ph18111622 - 27 Oct 2025
Viewed by 363
Abstract
Background: PARP inhibitors exhibit significant lethality in tumors with BRCA1/2 mutations or homologous recombination defects; however, their clinical application is limited by the rarity of BRCA1/2 mutations, complex resistance mechanisms, and limited efficacy of monotherapy. Objective: This study aimed to investigate [...] Read more.
Background: PARP inhibitors exhibit significant lethality in tumors with BRCA1/2 mutations or homologous recombination defects; however, their clinical application is limited by the rarity of BRCA1/2 mutations, complex resistance mechanisms, and limited efficacy of monotherapy. Objective: This study aimed to investigate how Niraparib induces dysregulation of sphingolipid metabolism—particularly upregulation of the key enzyme SPHK1—in triple-negative breast cancer (TNBC) cells, and to elucidate a novel super-enhancer (SE)-mediated mechanism of Niraparib resistance. We also applied AI-based virtual screening to identify compounds targeting key nodes and develop strategies for sensitizing TNBC to Niraparib. Results: Niraparib induced sphingolipid metabolic imbalance and significantly upregulated SPHK1 in TNBC. Multi-omics analyses revealed that SPHK1 is regulated by a super-enhancer. Mechanistically, Niraparib inhibited PARylation of the transcription factor SP1, enhancing its occupancy at the SE region and reactivating its transcriptional activity, thereby promoting SPHK1 expression. This process activated pro-survival signaling pathways and conferred niraparib resistance. AI-facilitated virtual screening identified the natural compound Echinatin as a potent SP1 inhibitor, which stably binds to SP1 and exhibits marked synergistic effects with Niraparib. While targeting downstream SPHK1 also provided therapeutic benefit by enhancing the anti-tumor efficacy of Niraparib, Echinatin demonstrates a superior advantage due to its more favorable toxicity profile. Conclusions: Niraparib induces resistance through the SP1-SE-SPHK1 axis, whereas Echinatin effectively reverses this mechanism by inhibiting the upstream regulator SP1, significantly potentiating the efficacy of Niraparib. This study reveals a novel molecular mechanism underlying Niraparib resistance and proposes a promising combination therapy strategy. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Graphical abstract

15 pages, 422 KB  
Article
Evaluating the Role of Liquid Biopsy to Detect Pathogenic Homologous Recombination Repair (HRR) Gene Alterations in Metastatic Prostate Cancer
by Soumaya Labidi, Belinda Jiao, Shirley Tam, Parvaneh Fallah, Aida Salehi, Raghu Rajan, Mona Alameldin, Fadi Brimo, William D. Foulkes, Andreas I. Papadakis, Nabodita Kaul, Alan Spatz, Cristiano Ferrario, Ramy R. Saleh and April A. N. Rose
Cancers 2025, 17(21), 3427; https://doi.org/10.3390/cancers17213427 - 25 Oct 2025
Viewed by 406
Abstract
Background: Metastatic prostate cancers frequently harbour pathogenic aberrations in Homologous Recombination Repair (HRR) genes that confer sensitivity to PARP inhibitors (PARPi). Therefore, accurate identification of all eligible patients is needed. The development of a circulating tumour DNA (ctDNA) testing alternative is promising as [...] Read more.
Background: Metastatic prostate cancers frequently harbour pathogenic aberrations in Homologous Recombination Repair (HRR) genes that confer sensitivity to PARP inhibitors (PARPi). Therefore, accurate identification of all eligible patients is needed. The development of a circulating tumour DNA (ctDNA) testing alternative is promising as genomic testing of archived tissue leads to a failure rate of up to 30–40% in prostate cancer. Methods: This was a bi-institutional retrospective cohort study of patients with metastatic prostate cancer treated at the Jewish General Hospital or the McGill University Health Center, Montreal, Canada, between 2021 and 2023. Molecular data and treatment information were abstracted from a chart review. Chi-square, Fisher’s exact test, and Mann–Whitney tests were used to assess differences between groups. Results: We identified 484 metastatic prostate cancer patients. Somatic and germline testing for HRR was performed in 55.4% (n = 268) and 20% (n = 97) patients, respectively. Somatic testing was performed on tissue (n = 192, 71.6%) or ctDNA from liquid biopsies (n = 18, 6.7%) or both (n = 58, 21.7%). Pathogenic somatic HRR alterations were detected in 48 patients (17.9%). BRCA2 was the most frequent (n = 17), followed by ATM (n = 11), then CHEK2 (n = 5). Amongst patients with germline testing, 13/97 (13.4%) had pathogenic alterations predicted to lead to deficient HRR, mostly BRCA2 (n = 9), and three had detectable BRCA2 in tissue. Dual testing modality (tissue+ctDNA) significantly enhanced the detection rate of HRR alterations 19/58 (32.7%) vs. 29/210 (13.8%) for single testing modality (tissue or ctDNA), p = 0.008. The rate of inconclusive results was significantly lower in dual testing modality 0/58 (0%) vs. 25/210 in single testing modality (11.9%), p = 0.003. Amongst the 14 patients who had discordant results between liquid and tissue tests, HRR abnormalities were more frequently identified in ctDNA (n = 11) vs. tissue (n = 3). Patients who had HRR deficiency detected only in ctDNA had older tissue samples (median 5.6 years) compared to those who had deficient HRR detected only in tissue (median 0.2 years; p = 0.14). Conclusions: These data highlight a potential role in implementing liquid biopsy—especially in patients who only have older archival tissue available or failed tissue testing—to improve the detection rate of deficient HRR. Our ongoing prospective study will further validate whether the addition of liquid biopsy can identify more patients who are eligible to receive precision therapies by increasing the rate of detection of HRR deficiency compared to routine tissue testing alone. Full article
(This article belongs to the Section Cancer Metastasis)
Show Figures

Figure 1

27 pages, 1763 KB  
Review
Molecular Mechanisms of the Ubiquitin-Specific Proteases (USPs) Family in Biliary Tract Cancer and Targeted Intervention Strategies
by Qian Cheng, Delin Ma, Shengmin Zheng, Jialing Hao, Gang Wang, Yanbin Ni and Jiye Zhu
Biomedicines 2025, 13(11), 2586; https://doi.org/10.3390/biomedicines13112586 - 23 Oct 2025
Viewed by 500
Abstract
Biliary tract carcinoma (BTC) is a group of highly heterogeneous malignancies arising from the biliary epithelium. Anatomically, BTC is categorized into gallbladder cancer (GBC) and cholangiocarcinoma (CCA), with the latter further subdivided into intrahepatic (iCCA), perihilar (pCCA), and distal cholangiocarcinoma (dCCA). Epidemiological studies [...] Read more.
Biliary tract carcinoma (BTC) is a group of highly heterogeneous malignancies arising from the biliary epithelium. Anatomically, BTC is categorized into gallbladder cancer (GBC) and cholangiocarcinoma (CCA), with the latter further subdivided into intrahepatic (iCCA), perihilar (pCCA), and distal cholangiocarcinoma (dCCA). Epidemiological studies reveal a dismal five-year survival rate of less than 20% for BTC patients, with limited responses to current chemotherapy regimens, underscoring the urgent need to unravel its complex molecular pathogenesis. Recent research has increasingly focused on the regulatory networks of post-translational modifications, particularly the ubiquitin-proteasome system (UPS), in tumorigenesis. As the largest subfamily of deubiquitinating enzymes (DUBs), ubiquitin-specific proteases (USPs) regulate the stability of key oncoproteins such as phosphatase and tensin homolog (PTEN) and c-Myc, playing pivotal roles in tumor cell proliferation, apoptosis evasion, invasion, and metastasis. This review systematically summarizes the differential expression profiles of USP family members (e.g., USP1, USP3, USP7, USP8, USP9X, USP21, and USP22) in BTC and their clinical significance, with a focus on elucidating how specific USPs regulate tumor progression through key substrates, including poly(ADP-ribose) polymerase 1 (PARP1), dynamin-1-like protein (DNM1L), and O-GlcNAc transferase (OGT). Furthermore, based on recent advances, we discuss the therapeutic potential of small-molecule USP inhibitors in BTC targeted therapy, providing a theoretical foundation for developing novel precision treatment strategies. Full article
(This article belongs to the Special Issue Ubiquitylation and Deubiquitylation in Health and Diseases)
Show Figures

Figure 1

21 pages, 609 KB  
Review
Artificial Intelligence Tools for Supporting Histopathologic and Molecular Characterization of Gynecological Cancers: A Review
by Aleksandra Asaturova, João Pinto, António Polonia, Evgeny Karpulevich, Xavier Mattias-Guiu and Catarina Eloy
J. Clin. Med. 2025, 14(21), 7465; https://doi.org/10.3390/jcm14217465 - 22 Oct 2025
Viewed by 473
Abstract
Background/Objectives: Accurate diagnosis, prognosis, and prediction of treatment response are essential in managing gynecologic cancers and maintaining patient quality of life. Computational pathology, powered by artificial intelligence (AI), offers a transformative opportunity for objective histopathological assessment. This review provides a comprehensive, user-oriented [...] Read more.
Background/Objectives: Accurate diagnosis, prognosis, and prediction of treatment response are essential in managing gynecologic cancers and maintaining patient quality of life. Computational pathology, powered by artificial intelligence (AI), offers a transformative opportunity for objective histopathological assessment. This review provides a comprehensive, user-oriented overview of existing AI tools for the characterization of gynecological cancers, critically evaluating their clinical applicability and identifying key challenges for future development. Methods: A systematic literature search was conducted in PubMed and Web of Science for studies published up to 2025. The search focused on AI tools developed for the diagnosis, prognosis, or treatment prediction of gynecologic cancers based on histopathological images. After applying selection criteria, 36 studies were included for in-depth analysis, covering ovarian, uterine, cervical, and other gynecological cancers. Studies on cytopathology and pure tumor detection were excluded. Results: Our analysis identified AI tools addressing critical clinical tasks, including histopathologic subtyping, grading, staging, molecular subtyping, and prediction of therapy response (e.g., to platinum-based chemotherapy or PARP inhibitors). The performance of these tools varied significantly. While some demonstrated high accuracy and promising results in internal validation, many were limited by a lack of external validation, potential biases from training data, and performance that is not yet sufficient for routine clinical use. Direct comparison between studies was often hindered by the use of non-standardized evaluation metrics and evolving disease classifications over the past decade. Conclusions: AI tools for gynecologic cancers represent a promising field with the potential to significantly support pathological practice. However, their current development is heterogeneous, and many tools lack the robustness and validation required for clinical integration. There is a pressing need to invest in the creation of clinically driven, interpretable, and accurate AI tools that are rigorously validated on large, multicenter cohorts. Future efforts should focus on standardizing evaluation metrics and addressing unmet diagnostic needs, such as the molecular subtyping of rare tumors, to ensure these technologies can reliably benefit patient care. Full article
Show Figures

Figure 1

14 pages, 1364 KB  
Systematic Review
Influence of Residual Disease on the Efficacy of PARP Inhibitors in Advanced Epithelial Ovarian Cancer: A Systematic Review and Meta Analysis
by Sekyoung Hwang, Ji Hyun Kim, Uisuk Kim, Hyeong In Ha, Sang-Yoon Park and Myong Cheol Lim
Cancers 2025, 17(20), 3365; https://doi.org/10.3390/cancers17203365 - 18 Oct 2025
Viewed by 708
Abstract
Objective: While PARP inhibitors (PARPi) improve progression-free survival (PFS) in advanced ovarian cancer, their efficacy across different surgical outcomes is unclear. We aimed to determine if the efficacy of PARPi maintenance therapy, as measured by PFS, is modified by postoperative residual disease (R0 [...] Read more.
Objective: While PARP inhibitors (PARPi) improve progression-free survival (PFS) in advanced ovarian cancer, their efficacy across different surgical outcomes is unclear. We aimed to determine if the efficacy of PARPi maintenance therapy, as measured by PFS, is modified by postoperative residual disease (R0 vs. R1/R2) in newly diagnosed advanced epithelial ovarian cancer. Methods: A systematic review and trial-level meta analysis of randomized controlled trials published through July 2025 was conducted. The primary endpoint was pooled hazard ratio (HR) for PFS, with subgroup analyses based on residual disease (R0 vs. R1/R2), clinical risk (higher risk vs. lower risk), and timing of surgery (primary cytoreductive surgery vs. interval cytoreductive surgery). Results: Six randomized controlled trials involving 3629 patients were included in this meta analysis. PARPi maintenance significantly improved PFS in both patients with no gross residual disease (R0) (HR 0.55, 95% CI 0.44–0.68, I2 = 64.2%) and those with macroscopic residual disease (R1/R2) (HR 0.51, 95% CI 0.40–0.65, I2 = 56.0%). The treatment effect did not differ significantly between these subgroups (p = 0.66). A numerically greater benefit was observed in lower-risk populations (HR 0.40, 95% CI 0.29–0.55, I2 = 0.9%) compared to higher-risk populations (HR 0.51, 95% CI 0.36–0.73, I2 = 78.5%, p = 0.30). The benefit was maintained irrespective of surgical timing, with similar pooled HRs for patients undergoing primary (HR 0.56, 95% CI 0.42–0.74, I2 = 72.3%) versus interval (HR 0.54, 95% CI 0.45–0.66, I2 = 44.2%) cytoreductive surgery. Conclusions: PARP inhibitor maintenance therapy provides a significant PFS benefit regardless of residual disease status, supporting its use in all eligible patients. Complete cytoreduction, however, remains crucial, as it provides the best foundation for achieving optimal long-term outcomes and maximizing the benefits of maintenance therapy. Full article
(This article belongs to the Special Issue PARP Inhibitors in Cancer Therapy)
Show Figures

Figure 1

16 pages, 1716 KB  
Review
Alternative Lengthening of Telomeres: A Prognostic Paradox in Cancer
by Ji-Yong Sung
Cells 2025, 14(20), 1613; https://doi.org/10.3390/cells14201613 - 17 Oct 2025
Viewed by 924
Abstract
Telomere maintenance enables unlimited cell proliferation by counteracting telomere erosion. While the majority of tumors activate telomerase, a significant subset—approximately 10–15%—utilizes alternative lengthening of telomeres (ALT), a recombination-based mechanism. ALT-positive cancers are classically associated with genomic instability, anaphase bridges, chromosomal rearrangements, and resistance [...] Read more.
Telomere maintenance enables unlimited cell proliferation by counteracting telomere erosion. While the majority of tumors activate telomerase, a significant subset—approximately 10–15%—utilizes alternative lengthening of telomeres (ALT), a recombination-based mechanism. ALT-positive cancers are classically associated with genomic instability, anaphase bridges, chromosomal rearrangements, and resistance to DNA-damaging therapies. This process is closely associated with genetic instability, which contributes to chromosomal rearrangements and tumor evolution. Consequently, ALT has traditionally been considered an adverse prognostic marker in aggressive malignancies such as osteosarcoma, pancreatic neuroendocrine tumors, and high-grade sarcomas. Paradoxically, recent evidence demonstrates that ALT positivity correlates with improved survival in glioblastoma (GBM) and chondrosarcoma, two tumor types that have historically been regarded as immune-cold and therapeutically intractable. This favorable outcome likely reflects a convergence of factors, including replication stress and DNA damage that impose a fitness cost in slow-growing or metabolically constrained tumors. Loss of ATRX/DAXX, while enabling ALT, further amplifies chromatin fragility, and ALT-mediated instability may paradoxically enhance immunogenicity within immune-quiescent microenvironments. Moreover, ALT-positive cells exhibit unique therapeutic vulnerabilities, particularly to ATR and PARP inhibitors. Together, these observations support a context-dependent model in which ALT functions as a double-edged sword, acting as a driver of malignant aggressiveness in rapidly proliferating cancers while serving as a relative liability in slower-growing, immune-cold tumors. Understanding this duality not only refines prognostic stratification but also opens opportunities for precision oncology. By integrating ALT-specific biomarkers into clinical workflows and exploiting ALT-related DNA repair dependencies, clinicians may transform a once uniformly negative prognostic factor into an actionable therapeutic target. Full article
Show Figures

Figure 1

17 pages, 3758 KB  
Article
IL-6 Blockade Enhances the Efficacy of CDK4/6 Inhibitor in BRCA1-Mutant Triple-Negative Breast Cancer Cells
by Li Pan, Changyou Shi, Joungil Choi and Jiayuh Lin
Cells 2025, 14(20), 1602; https://doi.org/10.3390/cells14201602 - 15 Oct 2025
Viewed by 661
Abstract
Breast cancer gene 1 (BRCA1) is a tumor suppressor gene essential for DNA repair, and its mutations are linked to aggressive breast cancers with poor prognosis. While poly (ADP-ribose) polymerase (PARP) inhibitors benefit some patients with BRCA1-mutant, human epidermal growth [...] Read more.
Breast cancer gene 1 (BRCA1) is a tumor suppressor gene essential for DNA repair, and its mutations are linked to aggressive breast cancers with poor prognosis. While poly (ADP-ribose) polymerase (PARP) inhibitors benefit some patients with BRCA1-mutant, human epidermal growth factor receptor 2 (HER2)-negative metastatic breast cancer, issues such as limited efficacy and drug resistance persist. This is especially critical for triple-negative breast cancer (TNBC), which lacks targeted therapies. Cyclin-dependent kinase 4/6 (CDK4/6) inhibitors such as abemaciclib—FDA-approved for estrogen receptor (ER)-positive/HER2-negative breast cancer—are emerging as potential treatments for TNBC. We evaluated abemaciclib in BRCA1-mutant TNBC cell lines (SUM149, HCC1937, and MDA-MB-436) and found them to be sensitive to the drug. However, treatment induced cellular senescence and Interleukin-6 (IL-6) secretion, which may promote drug resistance. To address this, we inhibited IL-6 signaling using bazedoxifene or glycoprotein 130 (GP130) siRNA, and both of which enhanced abemaciclib sensitivity. Combination treatment with bazedoxifene and abemaciclib synergistically inhibited cell migration and invasion, and induced apoptosis. In a mammary fat pad TNBC tumor model, the combination treatment significantly suppressed SUM149 tumor growth more than either agent alone. These findings support co-targeting IL-6 and CDK4/6 as a novel therapeutic strategy for BRCA1-mutant TNBC. Full article
Show Figures

Figure 1

22 pages, 642 KB  
Systematic Review
The Impact of Novel Therapies on Quality-of-Life in Triple-Negative Breast Cancer: A Systematic Review of Clinical Trials
by Banice Kamau, Maxim Shulimovich and Sinha Samridhi
Cancers 2025, 17(20), 3307; https://doi.org/10.3390/cancers17203307 - 13 Oct 2025
Viewed by 1033
Abstract
Background: Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype characterized by poor prognosis and limited therapeutic options. Chemotherapy regimens are associated with significant adverse effects negatively impacting patients’ quality of life (QoL). This systematic review aims to evaluate and compare QoL [...] Read more.
Background: Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype characterized by poor prognosis and limited therapeutic options. Chemotherapy regimens are associated with significant adverse effects negatively impacting patients’ quality of life (QoL). This systematic review aims to evaluate and compare QoL outcomes of patients with TNBC receiving novel therapies—including immunotherapy, antibody–drug conjugates, and targeted therapies—versus standard chemotherapy. Methods: We systematically reviewed randomized controlled trials (RCTs) published within the past 15 years, identified through comprehensive searches in PubMed, Google Scholar, Research4Life, and Elicit. Included studies involved FDA-approved novel therapies (pembrolizumab, atezolizumab, sacituzumab-govitecan, olaparib, and talazoparib) administered to TNBC patients, and assessed QoL using validated tools such as EORTC QLQ-C30. Observational studies, case reports, and non-standardized assessments were excluded. Results: Eight RCTs comprising 3929 patients met the inclusion criteria. Sacituzumab govitecan and PARP inhibitors (olaparib and talazoparib) significantly improved QoL, notably delaying deterioration across physical, emotional, and functional domains compared to standard chemotherapy. Conversely, immunotherapies (pembrolizumab, atezolizumab) showed non-significant trends toward QoL improvement, with effects varying by patient subgroup and disease stage. Interpretation was limited by study design differences, inconsistent compliance, and incomplete data reporting. Conclusions: Immunotherapy showed a neutral effect on quality of life, providing neither significant improvement nor additional decline. Olaparib was associated with a delayed deterioration in quality of life, showing a more favorable tolerability profile compared to chemotherapy. Talazoparib leads to clinically meaningful enhancements in quality of life, while sacituzumab govitecan effectively improves patient-reported outcomes relative to standard chemotherapy. Full article
(This article belongs to the Section Systematic Review or Meta-Analysis in Cancer Research)
Show Figures

Figure 1

31 pages, 2490 KB  
Review
Therapeutic Potential of Metal-Based and PARP Inhibitor Chemotherapy for BRCA1-Associated Triple-Negative Breast Cancer
by Adisorn Ratanaphan
Int. J. Mol. Sci. 2025, 26(20), 9881; https://doi.org/10.3390/ijms26209881 - 10 Oct 2025
Cited by 1 | Viewed by 1316
Abstract
Triple-negative breast cancer (TNBC) accounts for about 10–15% of all breast cancers and is an aggressive disease with a poor prognosis. There is currently no standard treatment regimen for TNBC patients; thus, chemotherapy remains the main treatment. Anthracycline- and taxane-based regimens are the [...] Read more.
Triple-negative breast cancer (TNBC) accounts for about 10–15% of all breast cancers and is an aggressive disease with a poor prognosis. There is currently no standard treatment regimen for TNBC patients; thus, chemotherapy remains the main treatment. Anthracycline- and taxane-based regimens are the most widely used in a clinical setting, either alone or in combination with other chemotherapeutic agents, including poly (ADP-ribose) polymerase (PARP) inhibitors and platinum drugs. Platinum drugs have been used particularly in patients with BRCA1-mutated TNBC. Preclinical and clinical trials revealed that the response to PARP inhibition was directly correlated to the sensitivity to platinum chemotherapies. Inhibition of PARP enzymes has been shown to specifically target BRCA1 dysfunctional cells. Therefore, targeting breast cancer cells that possess genetic alterations that are absent in normal cells could be attained by the exploitation of synthetic lethality for the discovery of other candidate metals, i.e., ruthenium-derived compounds, as next-generation drugs for the treatment of TNBC. This prospective approach provides new insight into alternative treatments for breast cancers with BRCA1-associated TNBC. Full article
(This article belongs to the Special Issue Toxicity of Metals, Metal-Based Drugs, and Microplastics)
Show Figures

Figure 1

Back to TopTop