2,604 results on '"Kinase Inhibitor"'
Search Results
2. Discovery of pyridine-based derivatives as FLT3 inhibitors for the treatment of acute myeloid leukemia
- Author
-
Chen, Beijing, Wang, Zhongyuan, Chen, Qi, Zhang, Ying, Wu, Shengfei, Zhang, Yu, Li, Aihong, Ouyang, Weiwei, Sima, Lijie, Li, Xiaoxu, Zhao, Dongsheng, Luo, Bilan, Wang, Jianta, Tang, Lei, Su, Xiaoming, and Liao, Weike
- Published
- 2025
- Full Text
- View/download PDF
3. A multi-model approach identifies ALW-II-41-27 as a promising therapy for osteoarthritis-associated inflammation and endochondral ossification
- Author
-
Ferrao Blanco, Mauricio N., Lesage, Raphaelle, Kops, Nicole, Fahy, Niamh, Bekedam, Fjodor T., Chavli, Athina, Bastiaansen-Jenniskens, Yvonne M., Geris, Liesbet, Chambers, Mark G., Pitsillides, Andrew A., Narcisi, Roberto, and van Osch, Gerjo J.V.M.
- Published
- 2024
- Full Text
- View/download PDF
4. Opportunities and challenges for targeting HPK1 in cancer immunotherapy
- Author
-
Xu, Jiamei, Li, Yingzhou, Chen, Xinyi, Yang, Junyi, Xia, Heye, Huang, Wenhai, and Zeng, Shenxin
- Published
- 2024
- Full Text
- View/download PDF
5. Design, synthesis, and in silico insights of novel N’-(2-oxoindolin-3-ylidene)piperidine-4-carbohydrazide derivatives as VEGFR-2 inhibitors
- Author
-
Eldehna, Wagdy M., Habib, Youmna A., Mahmoud, Abeer E., Barghash, Mohamed F., Elsayed, Zainab M., Elsawi, Ahmed E., Maklad, Raed M., Rashed, Mahmoud, Khalil, Amira, Hammad, Sherif F., Ali, Mamdouh M., and El Kerdawy, Ahmed M.
- Published
- 2024
- Full Text
- View/download PDF
6. Unveiling the anticancer potential of novel spirooxindole-tethered pyrazolopyridine derivatives
- Author
-
Eldehna, Wagdy M., Abdulla, Maha-Hamadien, Nafie, Mohamed S., Elsawi, Ahmed E., Ayman, Salsabil, Shahin, Mai I., Alhassan, Noura S., Zubaidi, Ahmad M., Ghabbour, Hazem A, Elaasser, Mahmoud, Al-Karmalawy, Ahmed A., and Abdel-Aziz, Hatem A.
- Published
- 2024
- Full Text
- View/download PDF
7. Identification of Thieno[3,2-d]pyrimidine derivatives as potent and selective Janus Kinase 1 inhibitors
- Author
-
Kim, Younghoon, Jeon, Eunhye, Ahn, Hyunwoo, Kang, Juhee, and Sim, Taebo
- Published
- 2025
- Full Text
- View/download PDF
8. Design, synthesis, and biological evaluation of a potent and orally bioavailable FGFRs inhibitor for fibrotic treatment
- Author
-
Yue, Lin, Tan, Zui, Wei, Wei, Liu, Hongyao, Xue, Taixiong, Su, Xingping, Wu, Xiuli, Xie, Yuting, Li, Peilin, Wang, Doudou, Liu, Zhihao, Gan, Cailing, and Ye, Tinghong
- Published
- 2025
- Full Text
- View/download PDF
9. Discovery of 4-amino-1,6-dihydro-7H-pyrrolo[2,3-d]pyridazin-7-one derivatives as potential receptor-interacting serine/threonine-protein kinase 1 (RIPK1) inhibitors
- Author
-
Zhang, Chufeng, Chen, Yulian, Li, Yong, Shi, Na, Teng, Yaxin, Li, Na, Tang, Minghai, Ma, Ziyan, Deng, Dexin, and Chen, Lijuan
- Published
- 2024
- Full Text
- View/download PDF
10. Discovery of potent Plasmodium falciparum protein kinase 6 (PfPK6) inhibitors with a type II inhibitor pharmacophore
- Author
-
Ong, Han Wee, Truong, Anna, Kwarcinski, Frank, de Silva, Chandi, Avalani, Krisha, Havener, Tammy M., Chirgwin, Michael, Galal, Kareem A., Willis, Caleb, Krämer, Andreas, Liu, Shubin, Knapp, Stefan, Derbyshire, Emily R., Zutshi, Reena, and Drewry, David H.
- Published
- 2023
- Full Text
- View/download PDF
11. Characterizing treatment resistance in muscle invasive bladder cancer using the chicken egg chorioallantoic membrane patient-derived xenograft model
- Author
-
Villanueva, Hugo, Wells, Gabrielle A., Miller, Malachi T., Villanueva, Mariana, Pathak, Ravi, Castro, Patricia, Ittmann, Michael M., Sikora, Andrew G., and Lerner, Seth P.
- Published
- 2022
- Full Text
- View/download PDF
12. Disrupted target binding with acryloyl group as potential Bcr-Abl/C-Src dual kinase inhibitor optimization strategies with maintained antitumor activity
- Author
-
Lin, Ching, Chiang, Hsin-Yi, Chen, Grace Shiahuy, Chern, Ji-Wang, and Yu, Chao-Wu
- Published
- 2025
- Full Text
- View/download PDF
13. Development of 3-indolyl substituted phenyl pyrazolo-carboxamide hybrids as potential type II VEGFR-2 inhibitors and in vitro cytotoxicity studies
- Author
-
Valapil, Durgesh Gurukkala, Devabattula, Geetanjali, Barahdia, Aman Singh, Godugu, Chandraiah, and Shankaraiah, Nagula
- Published
- 2025
- Full Text
- View/download PDF
14. Thieno[3,2‐b]pyridine: Attractive scaffold for highly selective inhibitors of underexplored protein kinases with variable binding mode.
- Author
-
Moyano, Paula Martín, Kubina, Tadeáš, Paruch, Štěpán Owen, Jarošková, Aneta, Novotný, Jan, Skočková, Veronika, Ovesná, Petra, Suchánková, Tereza, Prokofeva, Polina, Kuster, Bernhard, Šmída, Michal, Chaikuad, Apirat, Krämer, Andreas, Knapp, Stefan, Souček, Karel, and Paruch, Kamil
- Subjects
- *
PROTEIN kinases , *KINASE inhibitors , *PHARMACOPHORE , *PYRIDINE , *CANCER cells - Abstract
Protein kinases are key regulators of numerous biological processes and aberrant kinase activity can cause various diseases, particularly cancer. Herein, we report the identification of new series of highly selective kinase inhibitors based on the thieno[3,2‐b]pyridine scaffold. The weak interaction of the thieno[3,2‐b]pyridine core with the kinase hinge region allows for profoundly different binding modes all of which maintain high kinome‐wide selectivity, as illustrated by the isomers MU1464 and MU1668. Thus, this core structure provides a template of ATP‐competitive but not ATP‐mimetic inhibitors that are anchored at the kinase back pocket. Mapping the chemical space around the central thieno[3,2‐b]pyridine pharmacophore afforded highly selective inhibitors of the kinase Haspin, exemplified by the compound MU1920 that fulfils criteria for a quality chemical probe and is suitable for use in in vivo applications. However, despite the role of Haspin in mitosis, the inhibition of Haspin alone was not sufficient to elicit cytotoxic effect in cancer cells. The thieno[3,2‐b]pyridine scaffold can be used in a broader context, as a basis of inhibitors targeting other underexplored protein kinases, such as CDKLs. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
15. Discovery of novel FGFR4 inhibitors through a build-up fragment strategy.
- Author
-
Kim, Jihyung, Im, Chang Gyun, Oh, Kyujin, Lee, Ji Min, Al-Rubaye, Fatimah, and Min, Kyung Hoon
- Subjects
- *
HEPATOCELLULAR carcinoma , *SMALL molecules , *KINASE inhibitors , *WARHEADS , *FAMILIES - Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related death. FGFR4 has been implicated in HCC progression, making it a promising therapeutic target. We introduce an approach for identifying novel FGFR4 inhibitors by sequentially adding fragments to a common warhead unit. This strategy resulted in the discovery of a potent inhibitor, 4c, with an IC50 of 33 nM and high selectivity among members of the FGFR family. Although further optimisation is required, our approach demonstrated the potential for discovering potent FGFR4 inhibitors for HCC treatment, and provides a useful method for obtaining hit compounds from small fragments. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. Design and synthesis of doublecortin-like kinase 1 inhibitors and their bioactivity evaluation.
- Author
-
Pan, Pengming, Ji, Dengbo, Li, Zhongjun, and Meng, Xiangbao
- Subjects
- *
CANCER stem cells , *CHEMICAL synthesis , *KINASE inhibitors , *STRUCTURE-activity relationships , *DISEASE risk factors , *THREONINE - Abstract
Doublecortin-like kinase 1 (DCLK) is a microtubule-associated serine/threonine kinase that is upregulated in a wide range of cancers and is believed to be related to tumour growth and development. Upregulated DCLK1 has been used to identify patients at high risk of cancer progression and tumours with chemotherapy-resistance. Moreover, DCLK1 has been identified as a cancer stem cell (CSC) biomarker in various cancers, which has received considerable attention recently. Herein, a series of DCLK1 inhibitors were prepared based on the previously reported XMD8-92 structure. Among all the synthesised compounds, D1, D2, D6, D7, D8, D12, D14, and D15 showed higher DCLK1 inhibitory activities (IC50 40–74 nM) than XMD8-92 (IC50 161 nM). Compounds D1 and D2 were selective DCLK1 inhibitors as they showed a rather weak inhibitory effect on LRRK2. The antiproliferative activities of these compounds were also preliminarily evaluated. The structure-activity relationship revealed by our compounds provides useful guidance for the further development of DCLK1 inhibitors. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. Boronic Acid-Containing 3 H - pyrazolo[4,3- f ]quinoline Compounds as Dual CLK/ROCK Inhibitors with Anticancer Properties.
- Author
-
Dayal, Neetu, Chaudhuri, Riddhi, Yeboah, Kofi Simpa, Brauer, Nickolas R., and Sintim, Herman O.
- Subjects
- *
RHO-associated kinases , *CANCER cell growth , *CELL cycle , *RENAL cancer , *CELL migration - Abstract
Background: The protein kinases CLK and ROCK play key roles in cell growth and migration, respectively, and are potential anticancer targets. ROCK inhibitors have been approved by the FDA for various diseases and CLK inhibitors are currently being trialed in the clinic as anticancer agents. Compounds with polypharmacology are desired, especially in oncology, due to the potential for high efficacy as well as addressing resistance issues. In this report, we have identified and characterized novel, boron-containing dual CLK/ROCK inhibitors with promising anticancer properties. Methods: A library of boronic acid-based CLK/ROCKi was synthesized via Povarov/Doebner-type multicomponent reactions. Kinase inhibition screening and cancer cell viability assays were performed to identify the hit compounds. To gain insights into the probable binding modes of the compounds to the kinases, docking studies were performed. Cell cycle analysis, qPCR and immunoblotting were carried out to further characterize the mode(s) of action of the lead candidates. Results: At 25 nM, the top compounds HSD1400 and HSD1791 inhibited CLK1 and 2 and ROCK2 at greater than 70%. While HSD1400 also inhibited CLK4, the C1 methylated analog HSD1791 did not inhibit CLK4. Antitumor effects of the top compounds were evaluated and dose–response analysis indicated potent inhibition of renal cancer and leukemia cell growth. Immunoblotting results indicated that the top compounds induce DNA damage via upregulation of p-H2AX. Moreover, flow cytometry results demonstrated that the top compounds promote cell cycle arrest in the renal cancer cell line, Caki-1. qPCR and immunoblotting analysis upon HSD1791 dosing indicated suppression of cyclin D/Rb oncogenic pathway upon compound treatment. Conclusions: Novel boronic acid-containing pyrazolo[4,3-f]quinoline-based dual CLK/ROCK inhibitors were identified. The so-called "magic methylation" design approach was used to tune CLK selectivity. Additionally, the findings demonstrate potent in vitro anticancer activity of the lead candidates against renal cancer and leukemia. This adds to the growing list of boron-containing compounds that display biological activities. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. TPCA‐1 compound, inhibiting testis‐specific serine/threonine protein kinase 3 for potential male sterile in Bombyx mori.
- Author
-
Liu, Lianlian, Lu, Xiuping, Fan, Zeling, Deng, Jing, Zhang, Surui, Zhang, Lulu, and Zha, Xingfu
- Subjects
SMALL molecules ,RNA interference ,SILKWORMS ,PROTEIN kinases ,SMALL interfering RNA - Abstract
BACKGROUND: Protein kinases are a type of transferase enzyme that catalyze the phosphorylation of protein substrates, including receptor proteins. Testis‐specific serine/threonine kinases (TSSKs) are a highly conserved group of protein kinases found in various organisms. They play an essential role in male reproduction by influencing sperm development and function. RESULTS: In this study, we report on the characterization of BmTSSK3, a TSSK from the silkworm, Bombyx mori. We found that BmTSSK3 is specifically expressed in the testis and localized to the sperm flagella, particularly in the sperm tail cyst. Furthermore, we developed BmTSSK3 inhibitors through molecular docking and binding assays. Small molecules 5‐(4‐Fluorophenyl)‐2‐ureidothiophene‐3‐carboxamide (TPCA‐1) and Imidurea were identified to bind to BmTSSK3. Using site‐specific mutation technology, we identified amino acid residues R134 and S184 as crucial binding sites for small molecules. RNA interference assay and Western blot analysis showed that knockdown of BmTSSK3 significantly decreased histone 3 phosphorylation. To confirm the inhibitory effect of these small molecules, we treated silkworm testes with TPCA‐1 and observed a strong inhibitory effect. CONCLUSION: TPCA‐1 is an inhibitor of BmTSSK3, which raises its potential as a future candidate for male sterility of the silkworm. Thus, this study may offer a novel strategy for sterile silkworms as well as insects. © 2024 Society of Chemical Industry. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
19. Pharmacoproteogenomic approach identifies on-target kinase inhibitors for cancer drug repositioning: PHARMACOPROTEOGENOMIC APPROACH FOR CANCER DRUG REPOSITIONING: NOGUCHI ET AL.
- Author
-
Noguchi, Rei, Osaki, Julia, Ono, Takuya, Adachi, Yuki, Iwata, Shuhei, Yoshimatsu, Yuki, Sasaki, Kazuki, Kawai, Akira, and Kondo, Tadashi
- Abstract
Drug repositioning of approved drugs offers advantages over de novo drug development for a rare type of cancer. To efficiently identify on-target drugs from clinically successful kinase inhibitors in cancer drug repositioning, drug screening and molecular profiling of cell lines are essential to exclude off-targets. We developed a pharmacoproteogenomic approach to identify on-target kinase inhibitors, combining molecular profiling of genomic features and kinase activity, and drug screening of patient-derived cell lines. This study examined eight patient-derived giant cell tumor of the bone (GCTB) cell lines, all of which harbored a signature mutation of H3-3A but otherwise without recurrent copy number variants and mutations. Kinase activity profiles of 100 tyrosine kinases with a three-dimensional substrate peptide array revealed that nine kinases were highly activated. Pharmacological screening of 60 clinically used kinase inhibitors found that nine drugs directed at 29 kinases strongly suppressed cell viability. We regarded ABL1, EGFR, and LCK as on-target kinases; among the two corresponding on-target kinase inhibitors, osimertinib and ponatinib emerged as on-target drugs whose target kinases were significantly activated. The remaining 26 kinases and seven kinase inhibitors were excluded as off-targets. Our pharmacoproteomic approach enabled the identification of on-target kinase inhibitors that are useful for drug repositioning. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
20. A Systematic Review and Meta-Analysis of the Efficacy and Safety of Regorafenib in the Treatment of Metastatic Colorectal Cancer.
- Author
-
Liang, Bingjun, Tang, Ming, Huang, Chao, Yang, Yidian, He, Yue, Liao, Shengrong, and Shen, Weizeng
- Abstract
Background and Objective: Colorectal cancer (CRC) is a leading cause of cancer-related mortality worldwide. Despite advances in treatment, metastatic colorectal cancer (mCRC) remains a significant challenge due to its heterogeneity and resistance to therapy. Regorafenib, a multikinase inhibitor, can inhibit tumor progression through multiple mechanisms, thereby improving patient prognosis. It has emerged as a potential treatment option for mCRC patients who have progressed on standard therapies. This systematic review and meta-analysis aims to evaluate the efficacy and safety of Regorafenib in this patient population, synthesizing data from clinical trials to provide a comprehensive understanding of its role in mCRC treatment. Methods: A systematic literature search was conducted via the PubMed, Web of Science (WOS), and Embase databases from January 2012 to December 2024. Studies were included if they were randomized controlled trials (RCTs) or clinical trials that reported outcomes of regorafenib treatment in mCRC patients, including overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and disease control rate (DCR). Secondary outcomes included the incidence of serious adverse events (SAEs). OS refers to the length of time from the start of treatment until the death of the patient from any cause, while mortality specifically denotes the number of deaths occurring within the study period. Data were extracted by two independent reviewers using a standardized form. The meta-analysis was performed using RevMan 5.0 statistical software. Results: A total of 5,082 articles were retrieved, and ultimately, 9 eligible studies involving a total of 2,823 patients were included. All 9 included studies reported OS and PFS. In these mCRC patients, the dose of regorafenib was usually 160 mg daily. The meta results indicated that the OS of patients in the regorafenib group was significantly different [MD = 1.33, 95% CI (0.33, 2.33), P = 0.009]. Eight studies reported the ORR of the disease [OR = 1.13, 95% CI (0.73, 1.76), P = 0.57]. Five studies reported the DCR, and the DCR of patients in the regorafenib group was significantly different from that of patients in the control group [OR = 3.45, 95% CI (2.04, 5.84), P < 0.00001]. The incidence of SAEs (> grade 3) was reported in all 9 included studies [OR = 2.48, 95% CI (1.29, 4.73), P = 0.006]. Conclusion: In this systematic review of prospective trials, regorafenib resulted in improved OS with manageable adverse effects for patients with advanced mCRC. Still, considering the safety, future research should focus on investigating the dose optimization of regorafenib, as well as predictive biomarkers for therapeutic efficacy. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
21. Osteosarcoma cells depend on MCL-1 for survival, and osteosarcoma metastases respond to MCL-1 antagonism plus regorafenib in vivo
- Author
-
Yanhao Ji, Michael A. Harris, Lucas M. Newton, Tiffany J. Harris, W. Douglas Fairlie, Erinna F. Lee, and Christine J. Hawkins
- Subjects
Osteosarcoma ,Sarcoma ,BH3-mimetic ,MCL-1 ,BCL-xL ,Kinase inhibitor ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Osteosarcoma is the most common form of primary bone cancer, which primarily afflicts children and adolescents. Chemotherapy, consisting of doxorubicin, cisplatin and methotrexate (MAP) increased the 5-year osteosarcoma survival rate from 20% to approximately 60% by the 1980s. However, osteosarcoma survival rates have remained stagnant for several decades. Patients whose disease fails to respond to MAP receive second-line treatments such as etoposide and, in more recent years, the kinase inhibitor regorafenib. BCL-2 and its close relatives enforce cellular survival and have been implicated in the development and progression of various cancer types. BH3-mimetics antagonize pro-survival members of the BCL-2 family to directly stimulate apoptosis. These drugs have been proven to be efficacious in other cancer types, but their use in osteosarcoma has been relatively unexplored to date. We investigated the potential efficacy of BH3-mimetics against osteosarcoma cells in vitro and examined their cooperation with regorafenib in vivo. We demonstrated that osteosarcoma cell lines could be killed through inhibition of MCL-1 combined with BCL-2 or BCL-xL antagonism. Inhibition of MCL-1 also sensitized osteosarcoma cells to killing by second-line osteosarcoma treatments, particularly regorafenib. Importantly, we found that inhibition of MCL-1 with the BH3-mimetic S63845 combined with regorafenib significantly prolonged the survival of mice bearing pulmonary osteosarcoma metastases. Together, our results highlight the importance of MCL-1 in osteosarcoma cell survival and present a potential therapeutic avenue that may improve metastatic osteosarcoma patient outcomes.
- Published
- 2024
- Full Text
- View/download PDF
22. Targeting erbB Pathways in Breast Cancer: Dual Kinase Inhibition for Brain Metastasis and Prevention of p185HER2/Neu Tumor Development
- Author
-
Goel PN, Katsumata M, Qian W, Mathur S, Ji MQ, Samanta A, Grover P, Sgouros G, Chang JC, and Greene MI
- Subjects
breast cancer ,er121 ,kinase inhibitor ,neoadjuvant model ,tnbc ,brain metastasis ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Peeyush N Goel,1,2,* Makoto Katsumata,3,* Wei Qian,4 Sunil Mathur,4 Mei Q Ji,1 Arabinda Samanta,1 Payal Grover,1 George Sgouros,5 Jenny C Chang,4 Mark I Greene1 1Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104-6082, USA; 2Children’s Hospital of Philadelphia, Philadelphia, PA, 19104-6082, USA; 3Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA; 4Dr. Mary and Ron Neal Cancer Center, Houston Methodist Hospital, Houston, TX, USA; 5The Russell H Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA*These authors contributed equally to this workCorrespondence: Mark I Greene, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 252 John Morgan Building, 3620 hamilton Walk, Philadelphia, PA, 19104, USA, Tel +215-898-2847, Email greene@reo.med.upenn.eduBackground: Breast cancer predominantly affects women and poses challenges in the treatment of both local and advanced diseases. In a previous study, we reported the effectiveness of ER121, a structurally resolved small compound specifically designed to target human cancers expressing or overexpressing mutant EGFR and HER2.Purpose: The objective of this study is to assess the efficacy and toxicity of ER121 in metastatic and triple negative breast cancer (TNBC, HER2+) cells and tumor models. The Herceptin-resistant breast cancer cell line JIMT-1 was used in an in vivo tumor model, and MMTV-erbB2 (Fo5) transgenic mice models were used to evaluate the efficacy and safety of ER121 as neoadjuvant.Methods: ER121 treatment focusing on experimental brain metastasis in TNBC, HER2+ model, was quantified by total flux employing the In Vivo Imaging System (IVIS). We also compared the brain tissue from the treated and the controls groups. Additionally, ER121 was evaluated in JIMT-1, a Herceptin-resistant breast cancer cell line, both in vitro and in vivo tumor model. We also administered ER121 orally in neoadjuvant model with the MMTV-erbB2 (Fo5) transgenic mice, the survival rates were compared with the control group. Tumor-free survival of multiple treated groups were analyzed by Kaplan-Meier analysis employing the log-rank test with the Bonferroni correction using R Statistical Software.Results: In this study, we present findings indicating that ER121 treatment significantly attenuated breast tumor growth using a TNBC, HER2+ model, focusing on experimental brain metastasis, as quantified by total flux employing IVIS. These observations were further corroborated by analysis of brain tissue from the treatment group compared to controls. Data is presented as Mean ± S.D. statistical significance was calculated using Student t test (*p
- Published
- 2024
23. Regorafenib Combination Therapy in Advanced Hepatocellular Carcinoma: With or without Transarterial Chemoembolization.
- Author
-
Cao, Lei, Lu, Xiangyu, Chen, Haoqing, Yu, Xiang, Li, Jinze, Peng, Yi, Gu, Lu, Feng, Ji, Xie, Ping, and Liu, Yaben
- Subjects
- *
GROUP psychotherapy , *PROPENSITY score matching , *IMMUNE checkpoint inhibitors , *OVERALL survival , *HEPATOCELLULAR carcinoma , *CHEMOEMBOLIZATION - Abstract
The effectiveness and tolerability of triple therapy, which combines regorafenib, a programmed death 1 (PD-1) inhibitor, and transarterial chemoembolization (TACE), were compared to dual therapy consisting of regorafenib and a PD-1 inhibitor in patients with advanced hepatocellular carcinoma (HCC).Introduction: A retrospective analysis was conducted on patients with advanced HCC who underwent second-line therapy from March 2019 to June 2022 at multiple centers. Patients were stratified into two groups: dual therapy (comprising regorafenib and a PD-1 inhibitor) and triple therapy (consisting of regorafenib, a PD-1 inhibitor, and TACE). Propensity score matching (PSM) was used to control for potential confounding variables.Methods: After PSM, 112 eligible patients were included, with 56 in the triple therapy group and 56 in the dual therapy group. Median overall survival (OS) was significantly longer in the triple therapy group (15.4 vs. 8.9 months,Results: p < 0.001), as was median progression-free survival (6.8 vs. 3.3 months,p < 0.001). The objective response rate (37.5% vs. 5.4%,p < 0.001) and disease control rate (73.2% vs. 44.6%,p = 0.002) were significantly higher in the triple therapy group compared to the dual therapy group. The incidence and severity of adverse events were similar between the two groups. Triple therapy demonstrated superior survival benefits compared to dual therapy in patients with advanced HCC. Additionally, the safety profiles of the two treatment regimens were comparable. [ABSTRACT FROM AUTHOR]Conclusion: - Published
- 2024
- Full Text
- View/download PDF
24. Virtual Screening, Molecular Dynamics, and Mechanism Study of Homeodomain-Interacting Protein Kinase 2 Inhibitor in Renal Fibroblasts.
- Author
-
Hu, Xinlan, Wu, Yan, Ouyang, Hanyi, Wu, Jiayan, Yao, Mengmeng, Chen, Zhuo, and Li, Qianbin
- Subjects
- *
PROTEIN kinase inhibitors , *RENAL fibrosis , *MOLECULAR dynamics , *RATTUS norvegicus , *INHIBITION of cellular proliferation - Abstract
Background/Objectives: Homeodomain-interacting protein kinase 2 (HIPK2) is critically involved in the progression of renal fibrosis. This study aims to identify and characterize a novel HIPK2 inhibitor, CHR-6494, and investigate its therapeutic potential. Methods: Using structure-based virtual screening and molecular dynamics simulations, we identified CHR-6494 as a potent HIPK2 inhibitor with an IC50 of 0.97 μM. The effects of CHR-6494 on the phosphorylation of p53 in Normal Rattus norvegicus kidney cells (NRK-49F) induced by transforming growth factor-β (TGF-β) were assessed, along with its impact on TGF-β signaling and downstream profibrotic markers. Results: CHR-6494 significantly reduces p53 phosphorylation induced by TGF-β and enhances the interaction between HIPK2 and seven in absentia 2 (SIAH2), facilitating HIPK2 degradation via proteasomal pathways. Both CHR-6494 and Abemaciclib inhibit NRK-49F cell proliferation and migration induced by TGF-β, suppressing TGF-β/Smad3 signaling and decreasing profibrotic markers such as Fibronectin I (FN-I) Collagen I and α-smooth muscle actin (α-SMA). Additionally, these compounds inhibit nuclear factor kappa-B (NF-κB) signaling and reduce inflammatory cytokine expression. Conclusions: The study highlights the dual functionality of HIPK2 kinase inhibitors like CHR-6494 and Abemaciclib as promising therapeutic candidates for renal fibrosis and inflammation. The findings provide new insights into HIPK2 inhibition mechanisms and suggest pathways for the design of novel HIPK2 inhibitors in the future. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
25. Osteosarcoma cells depend on MCL-1 for survival, and osteosarcoma metastases respond to MCL-1 antagonism plus regorafenib in vivo.
- Author
-
Ji, Yanhao, Harris, Michael A., Newton, Lucas M., Harris, Tiffany J., Fairlie, W. Douglas, Lee, Erinna F., and Hawkins, Christine J.
- Subjects
BONE cancer ,OSTEOSARCOMA ,CELL survival ,KINASE inhibitors ,SURVIVAL rate - Abstract
Osteosarcoma is the most common form of primary bone cancer, which primarily afflicts children and adolescents. Chemotherapy, consisting of doxorubicin, cisplatin and methotrexate (MAP) increased the 5-year osteosarcoma survival rate from 20% to approximately 60% by the 1980s. However, osteosarcoma survival rates have remained stagnant for several decades. Patients whose disease fails to respond to MAP receive second-line treatments such as etoposide and, in more recent years, the kinase inhibitor regorafenib. BCL-2 and its close relatives enforce cellular survival and have been implicated in the development and progression of various cancer types. BH3-mimetics antagonize pro-survival members of the BCL-2 family to directly stimulate apoptosis. These drugs have been proven to be efficacious in other cancer types, but their use in osteosarcoma has been relatively unexplored to date. We investigated the potential efficacy of BH3-mimetics against osteosarcoma cells in vitro and examined their cooperation with regorafenib in vivo. We demonstrated that osteosarcoma cell lines could be killed through inhibition of MCL-1 combined with BCL-2 or BCL-x
L antagonism. Inhibition of MCL-1 also sensitized osteosarcoma cells to killing by second-line osteosarcoma treatments, particularly regorafenib. Importantly, we found that inhibition of MCL-1 with the BH3-mimetic S63845 combined with regorafenib significantly prolonged the survival of mice bearing pulmonary osteosarcoma metastases. Together, our results highlight the importance of MCL-1 in osteosarcoma cell survival and present a potential therapeutic avenue that may improve metastatic osteosarcoma patient outcomes. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
26. Efficacy and safety of ritlecitinib, an oral JAK3/TEC family kinase inhibitor, in adolescent and adult patients with alopecia totalis and alopecia universalis.
- Author
-
Mesinkovska, Natasha, King, Brett, Zhang, Xingqi, Guttman‐Yassky, Emma, Magnolo, Nina, Sinclair, Rodney, Mizuashi, Masato, Shapiro, Jerry, Peeva, Elena, Banerjee, Anindita, Takiya, Liza, Cox, Lori Ann, Wajsbrot, Dalia, Kerkmann, Urs, Law, Ernest, Wolk, Robert, and Schaefer, Gregor
- Abstract
This post‐hoc analysis of the ALLEGRO phase 2b/3 study (NCT03732807) evaluated the efficacy and safety of ritlecitinib, an oral Janus kinase 3/TEC family kinase inhibitor, in patients with alopecia totalis (AT) and alopecia universalis (AU). Patients aged ≥ 12 years with alopecia areata (AA) and ≥50% scalp hair loss received once‐daily ritlecitinib 50 or 30 mg (± 4‐week 200‐mg loading dose) or placebo for 24 weeks. In a subsequent 24‐week extension period, the ritlecitinib groups continued their doses and patients initially assigned to placebo switched to ritlecitinib (200/50 or 50 mg daily). In this analysis, clinician‐ and patient‐reported hair regrowth outcomes were assessed at weeks 24 and 48 in four AA subgroups: AT/AU, AT, AU, and non‐AT/AU. Safety was monitored throughout. Of the 718 randomized patients, 151 (21%) and 147 (20%) were defined as having AT or AU, respectively. At week 24, Severity of Alopecia Tool (SALT) score ≤20 (≤20% scalp hair loss) response rates were higher in the ritlecitinib‐treated AT/AU, AT, and AU groups (7%–14%, 7%–21%, and 4%–10%, respectively) vs the placebo group (0% in the AT/AU, AT, and AU groups). The proportions of patients with a SALT score of ≤20 increased through week 48 (AT/AU, 13%–31%; AT, 11%–27%; AU, 6%–41%). Additionally, at week 24, 25%–43%, 32%–42%, and 12%–50% of patients with AT/AU, AT, and AU, respectively, who received ritlecitinib achieved a moderately or greatly improved response based on the Patient Global Impression of Change scale. Response rates generally increased through week 48 and were similar across AA subgroups. In patients with AT/AU, ritlecitinib was well tolerated with a safety profile consistent with that of the overall AA population. Ritlecitinib demonstrated clinical efficacy, patient‐reported improvement, and an acceptable safety profile in patients with AT and AU through week 48. A plain language summary of this study is available at https://doi.org/10.25454/pfizer.figshare.26879161. Clinicaltrials.gov: NCT03732807. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
27. Screening and identification of host signaling pathways for alleviating influenza-induced production of pro-inflammatory cytokines, IP-10, IL-8, and MCP-1, using a U937 cell-based influenza model
- Author
-
Si Chen, Yang Yu, Yue Su, Xiaoqin Lian, Lefang Jiang, Zhuogang Li, Mingxin Zhang, Yarou Gao, Haonan Zhang, Xingjian Zhu, Jiaxin Ke, and Xulin Chen
- Subjects
influenza virus ,pro-inflammatory cytokine ,kinase inhibitor ,protein tyrosine kinase ,mitogen-activated protein kinase ,Janus kinase/signal transducers and activators of transcription ,Microbiology ,QR1-502 - Abstract
Influenza virus infection initiates an exaggerated inflammatory response, which may culminate in a fatal cytokine storm characterized by the excessive production of pro-inflammatory cytokines. Prior research indicates that IP-10, IL-8, and MCP-1, primarily produced by monocytes and macrophages, play a crucial role in influenza-induced inflammation. The lung injury from influenza virus infection can be mitigated by suppressing or inhibiting these cytokines through knockout, knockdown, or targeted intervention approaches. To identify the key host signaling pathways responsible for producing pro-inflammatory cytokines, we utilized a U937 cell model that secretes IP-10, IL-8, and MCP-1 in response to influenza infection. This model has been previously validated in our laboratory as an appropriate system for screening anti-inflammatory agents and potential drug targets. We conducted a screening assay employing an inhibitor library consisting of 2,138 compounds that target various known pathways and host factors. Our findings indicated that inhibitors targeting protein tyrosine kinases and mitogen-activated protein kinases demonstrated superior efficacy in suppressing cytokine production induced by influenza A virus infection compared to inhibitors aimed at other host factors. Notably, a substantial proportion of the identified hits capable of inhibiting the expression of all three cytokines in the secondary screening were classified as tyrosine kinase inhibitors. Validation experiments further established that Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathways, along with p38 MAPK and Raf–MEK–ERK pathways, are the principal regulators of pro-inflammatory cytokine expression in monocytes and macrophages. Moreover, our results suggest that TKIs present promising opportunities as novel therapeutic agents against influenza-induced pneumonia.
- Published
- 2025
- Full Text
- View/download PDF
28. Impact of the clinically approved BTK inhibitors on the conformation of full-length BTK and analysis of the development of BTK resistance mutations in chronic lymphocytic leukemia
- Author
-
Raji E Joseph, Thomas E Wales, Sandrine Jayne, Robert G Britton, D Bruce Fulton, John R Engen, Martin JS Dyer, and Amy H Andreotti
- Subjects
kinase inhibitor ,allostery ,resistance mutations ,BTK ,Bruton’s tyrosine kinase ,CLL ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Inhibition of Bruton’s tyrosine kinase (BTK) has proven to be highly effective in the treatment of B-cell malignancies such as chronic lymphocytic leukemia (CLL), autoimmune disorders, and multiple sclerosis. Since the approval of the first BTK inhibitor (BTKi), Ibrutinib, several other inhibitors including Acalabrutinib, Zanubrutinib, Tirabrutinib, and Pirtobrutinib have been clinically approved. All are covalent active site inhibitors, with the exception of the reversible active site inhibitor Pirtobrutinib. The large number of available inhibitors for the BTK target creates challenges in choosing the most appropriate BTKi for treatment. Side-by-side comparisons in CLL have shown that different inhibitors may differ in their treatment efficacy. Moreover, the nature of the resistance mutations that arise in patients appears to depend on the specific BTKi administered. We have previously shown that Ibrutinib binding to the kinase active site causes unanticipated long-range effects on the global conformation of BTK (Joseph et al., 2020). Here, we show that binding of each of the five approved BTKi to the kinase active site brings about distinct allosteric changes that alter the conformational equilibrium of full-length BTK. Additionally, we provide an explanation for the resistance mutation bias observed in CLL patients treated with different BTKi and characterize the mechanism of action of two common resistance mutations: BTK T474I and L528W.
- Published
- 2024
- Full Text
- View/download PDF
29. 243 - Rheumatoid Arthritis
- Author
-
McInnes, Iain B.
- Published
- 2024
- Full Text
- View/download PDF
30. Efficacy and safety of chiauranib in a combination therapy in platinum-resistant or refractory ovarian cancer: a multicenter, open-label, phase Ib and II study
- Author
-
Jin Li, Jihong Liu, Rutie Yin, Dongling Zou, Hong Zheng, Junning Cao, Zhendong Chen, Wei Sun, Yunong Gao, Songling Zhang, Linjuan Zeng, Ruifang An, Xianping Lu, Shuang Ye, and Xiaohua Wu
- Subjects
Chiauranib ,Ovarian cancer ,Kinase inhibitor ,Combo therapy ,Clinical Trial ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Platinum-resistant or refractory ovarian cancer is a highly lethal gynecologic disease with limited treatment options. Chiauranib is a novel small-molecule selective inhibitor, which could effectively target multiple pathways including Aurora B and CSF-1R to inhibit cell cycle process and improve anti-tumor immune function, as long as VEGF pathway for tumor extinction. Methods A phase II study was sequentially conducted after a phase Ib monotherapy study to evaluate the efficacy of chiauranib combined with chemotherapy. Chinese patients with recurrent ovarian cancer were enrolled. Eligible patients received chiauranib combined with a maximum of six cycles of chemotherapy: etoposide (CE group) or weekly-paclitaxel (CP group). Patients, who exhibited a complete or partial response, or stable disease following combo treatment, progressed to maintenance phase to receive chiauranib monotherapy. Primary endpoint was progression-free survival (PFS) according to RECIST v1.1. Results From November 2017 to March 2019, 25 patients were enrolled in a phase 1b study and a median PFS of 3.7 months (95% CI 1.8–NE) was achieved by chiauranib monotherapy. From July 2019 to December 2020, a total of 47 patients were enrolled in the phase II study. One CP patient did not receive the study drugs, and three patients withdrew before the first tumor assessment. Thus, 43 patients (CE group: 22 patients; CP group: 21 patients) were included in the evaluation. The median PFS was 5·4 months (95% CI 2·8–5·6) and 5·6 months (95% CI 3·4–7·0), respectively. Conclusions This was the first study to evaluate chiauranib, a novel multi-targeted kinase inhibitor in patients with ovarian cancer. The administration of chiauranib along with etoposide or weekly-paclitaxel significantly enhanced the efficacy with manageable adverse events. This warrants further clinical studies on this novel treatment. A phase III study is promising and ongoing. Trial registration ClinicaTrials.gov identifier: NCT03901118 (phase II) and NCT03166891 (phase Ib).
- Published
- 2024
- Full Text
- View/download PDF
31. Novel therapeutic agents in clinical trials: emerging approaches in cancer therapy
- Author
-
Deepak Chandra Joshi, Anurag Sharma, Sonima Prasad, Karishma Singh, Mayank Kumar, Kajal Sherawat, Hardeep Singh Tuli, and Madhu Gupta
- Subjects
Cancer ,Cancer therapy ,Checkpoint inhibitor ,Kinase inhibitor ,CAR-T cell therapy ,Cancer vaccines ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Novel therapeutic agents in clinical trials offer a paradigm shift in the approach to battling this prevalent and destructive disease, and the area of cancer therapy is on the precipice of a trans formative revolution. Despite the importance of tried-and-true cancer treatments like surgery, radiation, and chemotherapy, the disease continues to evolve and adapt, making new, more potent methods necessary. The field of cancer therapy is currently witnessing the emergence of a wide range of innovative approaches. Immunotherapy, including checkpoint inhibitors, CAR-T cell treatment, and cancer vaccines, utilizes the host’s immune system to selectively target and eradicate malignant cells while minimizing harm to normal tissue. The development of targeted medicines like kinase inhibitors and monoclonal antibodies has allowed for more targeted and less harmful approaches to treating cancer. With the help of genomics and molecular profiling, “precision medicine” customizes therapies to each patient’s unique genetic makeup to maximize therapeutic efficacy while minimizing unwanted side effects. Epigenetic therapies, metabolic interventions, radio-pharmaceuticals, and an increasing emphasis on combination therapy with synergistic effects further broaden the therapeutic landscape. Multiple-stage clinical trials are essential for determining the safety and efficacy of these novel drugs, allowing patients to gain access to novel treatments while also furthering scientific understanding. The future of cancer therapy is rife with promise, as the integration of artificial intelligence and big data has the potential to revolutionize early detection and prevention. Collaboration among researchers, and healthcare providers, and the active involvement of patients remain the bedrock of the ongoing battle against cancer. In conclusion, the dynamic and evolving landscape of cancer therapy provides hope for improved treatment outcomes, emphasizing a patient-centered, data-driven, and ethically grounded approach as we collectively strive towards a cancer-free world.
- Published
- 2024
- Full Text
- View/download PDF
32. Novel therapeutic agents in clinical trials: emerging approaches in cancer therapy.
- Author
-
Joshi, Deepak Chandra, Sharma, Anurag, Prasad, Sonima, Singh, Karishma, Kumar, Mayank, Sherawat, Kajal, Tuli, Hardeep Singh, and Gupta, Madhu
- Subjects
MEDICAL personnel ,CANCER treatment ,CANCER vaccines ,TREATMENT effectiveness ,DRUG efficacy - Abstract
Novel therapeutic agents in clinical trials offer a paradigm shift in the approach to battling this prevalent and destructive disease, and the area of cancer therapy is on the precipice of a trans formative revolution. Despite the importance of tried-and-true cancer treatments like surgery, radiation, and chemotherapy, the disease continues to evolve and adapt, making new, more potent methods necessary. The field of cancer therapy is currently witnessing the emergence of a wide range of innovative approaches. Immunotherapy, including checkpoint inhibitors, CAR-T cell treatment, and cancer vaccines, utilizes the host's immune system to selectively target and eradicate malignant cells while minimizing harm to normal tissue. The development of targeted medicines like kinase inhibitors and monoclonal antibodies has allowed for more targeted and less harmful approaches to treating cancer. With the help of genomics and molecular profiling, "precision medicine" customizes therapies to each patient's unique genetic makeup to maximize therapeutic efficacy while minimizing unwanted side effects. Epigenetic therapies, metabolic interventions, radio-pharmaceuticals, and an increasing emphasis on combination therapy with synergistic effects further broaden the therapeutic landscape. Multiple-stage clinical trials are essential for determining the safety and efficacy of these novel drugs, allowing patients to gain access to novel treatments while also furthering scientific understanding. The future of cancer therapy is rife with promise, as the integration of artificial intelligence and big data has the potential to revolutionize early detection and prevention. Collaboration among researchers, and healthcare providers, and the active involvement of patients remain the bedrock of the ongoing battle against cancer. In conclusion, the dynamic and evolving landscape of cancer therapy provides hope for improved treatment outcomes, emphasizing a patient-centered, data-driven, and ethically grounded approach as we collectively strive towards a cancer-free world. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
33. Efficacy and safety of chiauranib in a combination therapy in platinum-resistant or refractory ovarian cancer: a multicenter, open-label, phase Ib and II study.
- Author
-
Li, Jin, Liu, Jihong, Yin, Rutie, Zou, Dongling, Zheng, Hong, Cao, Junning, Chen, Zhendong, Sun, Wei, Gao, Yunong, Zhang, Songling, Zeng, Linjuan, An, Ruifang, Lu, Xianping, Ye, Shuang, and Wu, Xiaohua
- Subjects
CHINESE people ,PROGRESSION-free survival ,CANCER patients ,KINASE inhibitors ,THERAPEUTICS ,OVARIAN cancer ,PACLITAXEL - Abstract
Background: Platinum-resistant or refractory ovarian cancer is a highly lethal gynecologic disease with limited treatment options. Chiauranib is a novel small-molecule selective inhibitor, which could effectively target multiple pathways including Aurora B and CSF-1R to inhibit cell cycle process and improve anti-tumor immune function, as long as VEGF pathway for tumor extinction. Methods: A phase II study was sequentially conducted after a phase Ib monotherapy study to evaluate the efficacy of chiauranib combined with chemotherapy. Chinese patients with recurrent ovarian cancer were enrolled. Eligible patients received chiauranib combined with a maximum of six cycles of chemotherapy: etoposide (CE group) or weekly-paclitaxel (CP group). Patients, who exhibited a complete or partial response, or stable disease following combo treatment, progressed to maintenance phase to receive chiauranib monotherapy. Primary endpoint was progression-free survival (PFS) according to RECIST v1.1. Results: From November 2017 to March 2019, 25 patients were enrolled in a phase 1b study and a median PFS of 3.7 months (95% CI 1.8–NE) was achieved by chiauranib monotherapy. From July 2019 to December 2020, a total of 47 patients were enrolled in the phase II study. One CP patient did not receive the study drugs, and three patients withdrew before the first tumor assessment. Thus, 43 patients (CE group: 22 patients; CP group: 21 patients) were included in the evaluation. The median PFS was 5·4 months (95% CI 2·8–5·6) and 5·6 months (95% CI 3·4–7·0), respectively. Conclusions: This was the first study to evaluate chiauranib, a novel multi-targeted kinase inhibitor in patients with ovarian cancer. The administration of chiauranib along with etoposide or weekly-paclitaxel significantly enhanced the efficacy with manageable adverse events. This warrants further clinical studies on this novel treatment. A phase III study is promising and ongoing. Trial registration: ClinicaTrials.gov identifier: NCT03901118 (phase II) and NCT03166891 (phase Ib). [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
34. Identification of aryl hydrocarbon receptor allosteric antagonists from clinically approved drugs.
- Author
-
Mosa, Farag E. S., Alqahtani, Mohammed A., El‐Ghiaty, Mahmoud A., Dyck, Jason R. B., Barakat, Khaled, and El‐Kadi, Ayman O. S.
- Subjects
- *
ARYL hydrocarbon receptors , *TRANSCRIPTION factors , *HETERODIMERS , *BINDING sites , *PHARMACOPHORE - Abstract
The human aryl hydrocarbon receptor (AhR), a ligand‐dependent transcription factor, plays a pivotal role in a diverse array of pathways in biological and pathophysiological events. This position AhR as a promising target for both carcinogenesis and antitumor strategies. In this study we utilized computational modeling to screen and identify FDA‐approved drugs binding to the allosteric site between α2 of bHLH and PAS‐A domains of AhR, with the aim of inhibiting its canonical pathway activity. Our findings indicated that nilotinib effectively fits into the allosteric pocket and forms interactions with crucial residues F82, Y76, and Y137. Binding free energy value of nilotinib is the lowest among top hits and maintains stable within its pocket throughout entire (MD) simulations time. Nilotinib has also substantial interactions with F295 and Q383 when it binds to orthosteric site and activate AhR. Surprisingly, it does not influence AhR nuclear translocation in the presence of AhR agonists; instead, it hinders the formation of the functional AhR‐ARNT‐DNA heterodimer assembly, preventing the upregulation of regulated enzymes like CYP1A1. Importantly, nilotinib exhibits a dual impact on AhR, modulating AhR activity via the PAS‐B domain and working as a noncompetitive allosteric antagonist capable of blocking the canonical AhR signaling pathway in the presence of potent AhR agonists. These findings open a new avenue for the repositioning of nilotinib beyond its current application in diverse diseases mediated via AhR. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
35. Neratinib, a pan ERBB/HER inhibitor, restores sensitivity of PTEN-null, BRAFV600E melanoma to BRAF/MEK inhibition.
- Author
-
DuBose, Evan, Bevill, Samantha M., Mitchell, Dana K., Sciaky, Noah, Golitz, Brian T., Dixon, Shelley A. H., Rhodes, Steven D., Bear, James E., Johnson, Gary L., and Angus, Steven P.
- Subjects
BRAF genes ,GENE expression ,MELANOMA ,DRUG synergism ,RNA sequencing ,DACARBAZINE - Abstract
Introduction: Approximately 50% of melanomas harbor an activating BRAFV600E mutation. Standard of care involves a combination of inhibitors targeting mutant BRAF and MEK1/2, the substrate for BRAF in the MAPK pathway. PTEN loss-of-function mutations occur in ~40% of BRAFV600E melanomas, resulting in increased PI3K/AKT activity that enhances resistance to BRAF/MEK combination inhibitor therapy. Methods: To compare the response of PTEN null to PTEN wild-type cells in an isogenic background, CRISPR/Cas9 was used to knock out PTEN in a melanoma cell line that harbors a BRAFV600E mutation. RNA sequencing, functional kinome analysis, and drug synergy screening were employed in the context of BRAF/MEK inhibition. Results: RNA sequencing and functional kinome analysis revealed that the loss of PTEN led to an induction of FOXD3 and an increase in expression of the FOXD3 target gene, ERBB3/HER3. Inhibition of BRAF and MEK1/2 in PTEN null, BRAFV600E cells dramatically induced the expression of ERBB3/HER3 relative to wild-type cells. A synergy screen of epigenetic modifiers and kinase inhibitors in combination with BRAFi/MEKi revealed that the pan ERBB/HER inhibitor, neratinib, could reverse the resistance observed in PTEN null, BRAFV600E cells. Conclusions: The findings indicate that PTEN null BRAFV600E melanoma exhibits increased reliance on ERBB/HER signaling when treated with clinically approved BRAFi/MEKi combinations. Future studies are warranted to test neratinib reversal of BRAFi/MEKi resistance in patient melanomas expressing ERBB3/HER3 in combination with its dimerization partner ERBB2/HER2. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
36. Effective Radiosensitization of HNSCC Cell Lines by DNA-PKcs Inhibitor AZD7648 and PARP Inhibitors Talazoparib and Niraparib.
- Author
-
Mentzel, Jacob, Hildebrand, Laura S., Kuhlmann, Lukas, Fietkau, Rainer, and Distel, Luitpold V.
- Subjects
- *
CELL lines , *POLY(ADP-ribose) polymerase , *CELL cycle , *IONIZING radiation , *DNA repair - Abstract
(1) Head and neck squamous cell carcinoma (HNSCC) is common, while treatment is difficult, and mortality is high. Kinase inhibitors are promising to enhance the effects of radiotherapy. We compared the effects of the PARP inhibitors talazoparib and niraparib and that of the DNA-PKcs inhibitor AZD7648, combined with ionizing radiation. (2) Seven HNSCC cell lines, including Cal33, CLS-354, Detroit 562, HSC4, RPMI2650 (HPV-negative), UD-SCC-2 and UM-SCC-47 (HPV-positive), and two healthy fibroblast cell lines, SBLF8 and SBLF9, were studied. Flow cytometry was used to analyze apoptosis and necrosis induction (AnnexinV/7AAD) and cell cycle distribution (Hoechst). Cell inactivation was studied by the colony-forming assay. (3) AZD7648 had the strongest effects, radiosensitizing all HNSCC cell lines, almost always in a supra-additive manner. Talazoparib and niraparib were effective in both HPV-positive cell lines but only consistently in one and two HPV-negative cell lines, respectively. Healthy fibroblasts were not affected by any combined treatment in apoptosis and necrosis induction or G2/M-phase arrest. AZD7648 alone was not toxic to healthy fibroblasts, while the combination with ionizing radiation reduced clonogenicity. (4) In conclusion, talazoparib, niraparib and, most potently, AZD7648 could improve radiation therapy in HNSCC. Healthy fibroblasts tolerated AZD7648 alone extremely well, but irradiation-induced effects might occur. Our results justify in vivo studies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
37. In silico identification of a novel Cdc2‐like kinase 2 (CLK2) inhibitor in triple negative breast cancer.
- Author
-
Huang, Cheng‐Chiao, Hsu, Chia‐Ming, Chao, Min‐Wu, Hsu, Kai‐Cheng, Lin, Tony Eight, Yen, Shih‐Chung, Tu, Huang‐Ju, and Pan, Shiow‐Lin
- Abstract
Dysregulation of RNA splicing processes is intricately linked to tumorigenesis in various cancers, especially breast cancer. Cdc2‐like kinase 2 (CLK2), an oncogenic RNA‐splicing kinase pivotal in breast cancer, plays a significant role, particularly in the context of triple‐negative breast cancer (TNBC), a subtype marked by substantial medical challenges due to its low survival rates. In this study, we employed a structure‐based virtual screening (SBVS) method to identify potential CLK2 inhibitors with novel chemical structures for treating TNBC. Compound 670551 emerged as a novel CLK2 inhibitor with a 50% inhibitory concentration (IC50) value of 619.7 nM. Importantly, Compound 670551 exhibited high selectivity for CLK2 over other protein kinases. Functionally, this compound significantly reduced the survival and proliferation of TNBC cells. Results from a cell‐based assay demonstrated that this inhibitor led to a decrease in RNA splicing proteins, such as SRSF4 and SRSF6, resulting in cell apoptosis. In summary, we identified a novel CLK2 inhibitor as a promising potential treatment for TNBC therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
38. Comprehensive multi-omics analysis elucidates colchicine-induced toxicity mechanisms and unveils the therapeutic potential of MLN4924 and kinase inhibitors
- Author
-
Zhai, Lin-hui, Jia, Xing-long, Chen, Yu-lu, Liu, Mu-yin, Zhang, Jing-dan, Ma, Shao-jie, Wang, Xiu-jun, Cheng, Wen-hao, He, Jing-liang, Zhou, Jiao-jiao, Zuo, Ling-yi, Zhang, Mei-qi, Yuan, Qing, Xu, Meng-han, Ji, Jing, Tan, Min-jia, and Liu, Bin
- Published
- 2024
- Full Text
- View/download PDF
39. Tailored management of advanced thyroid cancer patients treated with lenvatinib or vandetanib: the role of a multimodal approach
- Author
-
Nervo, Alice, Ferrari, Matteo, Vaccaro, Elisa, Migliore, Enrica, Gruosso, Giovanni, Roux, Anna, Piovesan, Alessandro, and Arvat, Emanuela
- Published
- 2024
- Full Text
- View/download PDF
40. First effectiveness data of lenvatinib and pembrolizumab as first-line therapy in advanced anaplastic thyroid cancer: a retrospective cohort study
- Author
-
Soll, Dominik, Bischoff, Philip, Frisch, Anne, Jensen, Marie, Karadeniz, Zehra, Mogl, Martina T., Horst, David, Penzkofer, Tobias, Spranger, Joachim, Keilholz, Ulrich, and Mai, Knut
- Published
- 2024
- Full Text
- View/download PDF
41. Discovery of dual rho-associated protein kinase 1 (ROCK1)/apoptosis signal–regulating kinase 1 (ASK1) inhibitors as a novel approach for non-alcoholic steatohepatitis (NASH) treatment
- Author
-
Zaky, Yara A., Rashad, Mai W., Zaater, Marwa A., and El Kerdawy, Ahmed M.
- Published
- 2024
- Full Text
- View/download PDF
42. Discovery and Anticancer Screening of Novel Oxindole-Based Derivative Bearing Pyridyl Group as Potent and Selective Dual FLT3/CDK2 Kinase Inhibitor.
- Author
-
Soudi, Aya, Bender, Onur, Celik, Ismail, El-Hafeez, Amer Ali Abd, Dogan, Rumeysa, Atalay, Arzu, Elkaeed, Eslam B., Alsfouk, Aisha A., Abdelhafez, Elshimaa M. N., Aly, Omar M., Sippl, Wolfgang, and Ali, Taha F. S.
- Subjects
- *
MEDICAL screening , *PROTEIN kinases , *KINASE inhibitors , *COLON cancer , *CYCLIN-dependent kinases , *KINASES , *PROTEIN-tyrosine kinases - Abstract
Protein kinases regulate cellular activities and make up over 60% of oncoproteins and proto-oncoproteins. Among these kinases, FLT3 is a member of class III receptor tyrosine kinase family which is abundantly expressed in individuals with acute leukemia. Our previous oxindole-based hit has a particular affinity toward FLT3 (IC50 = 2.49 μM) and has demonstrated selectivity towards FLT3 ITD-mutated MV4-11 AML cells, with an IC50 of 4.3 μM. By utilizing the scaffold of the previous hit, sixteen new compounds were synthesized and screened against NCI-60 human cancer cell lines. This leads to the discovery of a potent antiproliferative compound, namely 5l, with an average GI50 value against leukemia and colon cancer subpanels equalling 3.39 and 5.97 µM, respectively. Screening against a specific set of 10 kinases that are associated with carcinogenesis indicates that compound 5l has a potent FLT3 inhibition (IC50 = 36.21 ± 1.07 nM). Remarkably, compound 5l was three times more effective as a CDK2 inhibitor (IC50 = 8.17 ± 0.32 nM) compared to sunitinib (IC50 = 27.90 ± 1.80 nM). Compound 5l was further analyzed by means of docking and molecular dynamics simulation for CDK2 and FLT3 active sites which provided a rational for the observed strong inhibition of kinases. These results suggest a novel structural scaffold candidate that simultaneously inhibits CDK2 and FLT3 and gives encouragement for further development as a potential therapeutic for leukemia and colon cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
43. 2-Desaza-annomontine (C81) impedes angiogenesis through reduced VEGFR2 expression derived from inhibition of CDC2-like kinases.
- Author
-
Zech, T. J., Wolf, A., Hector, M., Bischoff-Kont, I., Krishnathas, G. M., Kuntschar, S., Schmid, T., Bracher, F., Langmann, T., and Fürst, R.
- Subjects
GENE expression ,MACULAR degeneration ,VASCULAR endothelial growth factors ,ALTERNATIVE RNA splicing ,NEOVASCULARIZATION - Abstract
Angiogenesis is a crucial process in the progression of various pathologies, like solid tumors, wet age-related macular degeneration, and chronic inflammation. Current anti-angiogenic treatments still have major drawbacks like limited efficacy in diseases that also rely on inflammation. Therefore, new anti-angiogenic approaches are sorely needed, and simultaneous inhibition of angiogenesis and inflammation is desirable. Here, we show that 2-desaza-annomontine (C81), a derivative of the plant alkaloid annomontine previously shown to inhibit endothelial inflammation, impedes angiogenesis by inhibiting CDC2-like kinases (CLKs) and WNT/β-catenin signaling. C81 reduced choroidal neovascularization in a laser-induced murine in vivo model, inhibited sprouting from vascular endothelial growth factor A (VEGF-A)-activated murine aortic rings ex vivo, and reduced angiogenesis-related activities of endothelial cells in multiple functional assays. This was largely phenocopied by CLK inhibitors and knockdowns, but not by inhibitors of the other known targets of C81. Mechanistically, CLK inhibition reduced VEGF receptor 2 (VEGFR2) mRNA and protein expression as well as downstream signaling. This was partly caused by a reduction of WNT/β-catenin pathway activity, as activating the pathway induced, while β-catenin knockdown impeded VEGFR2 expression. Surprisingly, alternative splicing of VEGFR2 was not detected. In summary, C81 and other CLK inhibitors could be promising compounds in the treatment of diseases that depend on angiogenesis and inflammation due to their impairment of both processes. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
44. First-In-Human Phase I Study of Tinengotinib (TT-00420), a Multiple Kinase Inhibitor, as a Single Agent in Patients With Advanced Solid Tumors.
- Author
-
Piha-Paul, Sarina A, Xu, Binghe, Dumbrava, Ecaterina E, Fu, Siqing, Karp, Daniel D, Meric-Bernstam, Funda, Hong, David S, Rodon, Jordi A, Tsimberidou, Apostolia M, Raghav, Kanwal, Ajani, Jaffer A, Conley, Anthony P, Mott, Frank, Fan, Ying, Fan, Jean, Peng, Peng, Wang, Hui, Ni, Shumao, Sun, Caixia, and Qiang, Xiaoyan
- Subjects
PROTEIN kinase inhibitors ,DRUG toxicity ,CASTRATION-resistant prostate cancer ,PATIENT safety ,RESEARCH funding ,INVESTIGATIONAL drugs ,PROTEIN-tyrosine kinase inhibitors ,CLINICAL trials ,CANCER patients ,DRUG dosage ,DESCRIPTIVE statistics ,JANUS kinases ,DRUG efficacy ,FIBROBLAST growth factors ,TUMORS ,NEUROTRANSMITTER uptake inhibitors ,GENETIC mutation ,COMPARATIVE studies ,EPIDERMAL growth factor receptors ,PHARMACODYNAMICS ,CHEMICAL inhibitors - Abstract
Purpose This first-in-human phase I dose-escalation study evaluated the safety, pharmacokinetics, and efficacy of tinengotinib (TT-00420), a multi-kinase inhibitor targeting fibroblast growth factor receptors 1-3 (FGFRs 1-3), Janus kinase 1/2, vascular endothelial growth factor receptors, and Aurora A/B, in patients with advanced solid tumors. Patients and Methods Patients received tinengotinib orally daily in 28-day cycles. Dose escalation was guided by Bayesian modeling using escalation with overdose control. The primary objective was to assess dose-limiting toxicities (DLTs), maximum tolerated dose (MTD), and dose recommended for dose expansion (DRDE). Secondary objectives included pharmacokinetics and efficacy. Results Forty-eight patients were enrolled (dose escalation, n = 40; dose expansion, n = 8). MTD was not reached; DRDE was 12 mg daily. DLTs were palmar-plantar erythrodysesthesia syndrome (8 mg, n = 1) and hypertension (15 mg, n = 2). The most common treatment-related adverse event was hypertension (50.0%). In 43 response-evaluable patients, 13 (30.2%) achieved partial response (PR; n = 7) or stable disease (SD) ≥ 24 weeks (n = 6), including 4/11 (36.4%) with FGFR2 mutations/fusions and cholangiocarcinoma (PR n = 3; SD ≥ 24 weeks n = 1), 3/3 (100.0%) with hormone receptor (HR)-positive/HER2-negative breast cancer (PR n = 2; SD ≥ 24 weeks n = 1), 2/5 (40.0%) with triple-negative breast cancer (TNBC; PR n = 1; SD ≥ 24 weeks n = 1), and 1/1 (100.0%) with castrate-resistant prostate cancer (CRPC; PR). Four of 12 patients (33.3%; HR-positive/HER2-negative breast cancer, TNBC, prostate cancer, and cholangiocarcinoma) treated at DRDE had PRs. Tinengotinib's half-life was 28-34 hours. Conclusions Tinengotinib was well tolerated with favorable pharmacokinetic characteristics. Preliminary findings indicated potential clinical benefit in FGFR inhibitor-refractory cholangiocarcinoma, HER2-negative breast cancer (including TNBC), and CRPC. Continued evaluation of tinengotinib is warranted in phase II trials. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
45. Crystal structure of (S)-5-(3-acetyl-5-chloro-2-ethoxy-6-fluorophenyl)-2-oxazolidinone.
- Author
-
Li, Victor, Yap, Glenn P. A., and Chaoying Ni
- Subjects
- *
CRYSTAL structure , *HYDROGEN bonding , *ANTINEOPLASTIC agents , *KINASE inhibitors - Abstract
The structure of (S)-5-(3-acetyl-5-chloro-2-ethoxy-6-fluorophenyl)-2-oxazolidinone, C13H13ClFNO4, at 100 K has monoclinic (P21) symmetry. The compound has a polymeric structure propagated by a screw axis parallel to the b axis with N--H⋯O hydrogen bonding. It is of interest with respect to efforts in the synthesis of a candidate anticancer drug, parsaclisib. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
46. Synthesis of 5-Benzylamino and 5-Alkylamino-Substituted Pyrimido[4,5-c]quinoline Derivatives as CSNK2A Inhibitors with Antiviral Activity.
- Author
-
Asressu, Kesatebrhan Haile, Smith, Jeffery L., Dickmander, Rebekah J., Moorman, Nathaniel J., Wellnitz, James, Popov, Konstantin I., Axtman, Alison D., and Willson, Timothy M.
- Subjects
- *
CONFORMATIONAL analysis , *HEPATITIS A virus , *QUINOLINE derivatives , *CHEMICAL derivatives , *VIRAL hepatitis , *HEPATITIS viruses - Abstract
A series of 5-benzylamine-substituted pyrimido[4,5-c]quinoline derivatives of the CSNK2A chemical probe SGC-CK2-2 were synthesized with the goal of improving kinase inhibitor cellular potency and antiviral phenotypic activity while maintaining aqueous solubility. Among the range of analogs, those bearing electron-withdrawing (4c and 4g) or donating (4f) substituents on the benzyl ring as well as introduction of non-aromatic groups such as the cyclohexylmethyl (4t) were shown to maintain CSNK2A activity. The CSNK2A activity was also retained with N-methylation of SGC-CK2-2, but α-methyl substitution of the benzyl substituent led to a 10-fold reduction in potency. CSNK2A inhibition potency was restored with indene-based compound 4af, with activity residing in the S-enantiomer (4ag). Analogs with the highest CSNK2A potency showed good activity for inhibition of Mouse Hepatitis Virus (MHV) replication. Conformational analysis indicated that analogs with the best CSNK2A inhibition (4t, 4ac, and 4af) exhibited smaller differences between their ground state conformation and their predicted binding pose. Analogs with reduced activity (4ad, 4ae, and 4ai) required more substantial conformational changes from their ground state within the CSNK2A protein pocket. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
47. Differentiated thyroid carcinoma: what the nonspecialists needs to know
- Author
-
Ana O. Hoff, Aline Lauda Freitas Chaves, Thiago Bueno de Oliveira, Helton Estrela Ramos, Gustavo Cancela Penna, Lucas Vieira dos Santos, Ana Luiza Maia, Daniel Oliveira Brito, and Franco Pelissari Vizzotto
- Subjects
Thyroid cancer ,differentiated thyroid carcinoma ,kinase inhibitor ,lenvatinib ,sorafenib ,Medicine ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
ABSTRACT Differentiated thyroid carcinoma (DTC) accounts for most cases of thyroid cancer, and the heterogeneity of DTC requires that management decisions be taken by a multidisciplinary team involving endocrinologists, head and neck surgeons, nuclear medicine physicians, pathologists, radiologists, radiation oncologists, and medical oncologists. It is important for nonspecialists to recognize and refer patients with DTC who will benefit from a specialized approach. Recent advances in knowledge and changes in management of DTC call for the need to raise awareness on the part of these nonspecialist physicians, including general endocrinologists and medical oncologists at large. We provide an overview of diagnostic and therapeutic principles in DTC, especially those that bear direct implication on day-to-day management of these patients by generalists. Patients with DTC may be broadly categorized as having localized, locally persistent/recurrent, or metastatic disease. Current recommendations for DTC include a three-tiered system that classifies patients with localized disease into low, intermediate, or high risk of persistent or recurrent disease. Risk stratification should be performed at baseline and repeated on an ongoing basis, depending on clinical evolution. One of the overarching goals in the management of DTC is the need to personalize treatment by tailoring its modality and intensity according to ongoing prognostic stratification, evolving knowledge about the disease, and patient characteristics and preference. In metastatic disease that is refractory to radioactive iodine, thyroid tumors are being reclassified into molecular subtypes that better reflect their biological properties and for which molecular alterations can be targeted with specific agents.
- Published
- 2024
- Full Text
- View/download PDF
48. Transfer learning enhanced graph neural network for aldehyde oxidase metabolism prediction and its experimental application
- Author
-
Jiacheng Xiong, Rongrong Cui, Zhaojun Li, Wei Zhang, Runze Zhang, Zunyun Fu, Xiaohong Liu, Zhenghao Li, Kaixian Chen, and Mingyue Zheng
- Subjects
Drug metabolism ,Aldehyde oxidase ,Transfer learning ,Graph neural network ,Kinase inhibitor ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Aldehyde oxidase (AOX) is a molybdoenzyme that is primarily expressed in the liver and is involved in the metabolism of drugs and other xenobiotics. AOX-mediated metabolism can result in unexpected outcomes, such as the production of toxic metabolites and high metabolic clearance, which can lead to the clinical failure of novel therapeutic agents. Computational models can assist medicinal chemists in rapidly evaluating the AOX metabolic risk of compounds during the early phases of drug discovery and provide valuable clues for manipulating AOX-mediated metabolism liability. In this study, we developed a novel graph neural network called AOMP for predicting AOX-mediated metabolism. AOMP integrated the tasks of metabolic substrate/non-substrate classification and metabolic site prediction, while utilizing transfer learning from 13C nuclear magnetic resonance data to enhance its performance on both tasks. AOMP significantly outperformed the benchmark methods in both cross-validation and external testing. Using AOMP, we systematically assessed the AOX-mediated metabolism of common fragments in kinase inhibitors and successfully identified four new scaffolds with AOX metabolism liability, which were validated through in vitro experiments. Furthermore, for the convenience of the community, we established the first online service for AOX metabolism prediction based on AOMP, which is freely available at https://aomp.alphama.com.cn.
- Published
- 2024
- Full Text
- View/download PDF
49. Allosteric coupling asymmetry mediates paradoxical activation of BRAF by type II inhibitors
- Author
-
Damien M Rasmussen, Manny M Semonis, Joseph T Greene, Joseph M Muretta, Andrew R Thompson, Silvia Toledo Ramos, David D Thomas, William CK Pomerantz, Tanya S Freedman, and Nicholas M Levinson
- Subjects
BRAF ,kinase inhibitor ,paradoxical activation ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
The type II class of RAF inhibitors currently in clinical trials paradoxically activate BRAF at subsaturating concentrations. Activation is mediated by induction of BRAF dimers, but why activation rather than inhibition occurs remains unclear. Using biophysical methods tracking BRAF dimerization and conformation, we built an allosteric model of inhibitor-induced dimerization that resolves the allosteric contributions of inhibitor binding to the two active sites of the dimer, revealing key differences between type I and type II RAF inhibitors. For type II inhibitors the allosteric coupling between inhibitor binding and BRAF dimerization is distributed asymmetrically across the two dimer binding sites, with binding to the first site dominating the allostery. This asymmetry results in efficient and selective induction of dimers with one inhibited and one catalytically active subunit. Our allosteric models quantitatively account for paradoxical activation data measured for 11 RAF inhibitors. Unlike type II inhibitors, type I inhibitors lack allosteric asymmetry and do not activate BRAF homodimers. Finally, NMR data reveal that BRAF homodimers are dynamically asymmetric with only one of the subunits locked in the active αC-in state. This provides a structural mechanism for how binding of only a single αC-in inhibitor molecule can induce potent BRAF dimerization and activation.
- Published
- 2024
- Full Text
- View/download PDF
50. Neratinib, a pan ERBB/HER inhibitor, restores sensitivity of PTEN-null, BRAFV600E melanoma to BRAF/MEK inhibition
- Author
-
Evan DuBose, Samantha M. Bevill, Dana K. Mitchell, Noah Sciaky, Brian T. Golitz, Shelley A. H. Dixon, Steven D. Rhodes, James E. Bear, Gary L. Johnson, and Steven P. Angus
- Subjects
melanoma ,kinase inhibitor ,drug synergy ,ERBB3 ,resistance ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
IntroductionApproximately 50% of melanomas harbor an activating BRAFV600E mutation. Standard of care involves a combination of inhibitors targeting mutant BRAF and MEK1/2, the substrate for BRAF in the MAPK pathway. PTEN loss-of-function mutations occur in ~40% of BRAFV600E melanomas, resulting in increased PI3K/AKT activity that enhances resistance to BRAF/MEK combination inhibitor therapy.MethodsTo compare the response of PTEN null to PTEN wild-type cells in an isogenic background, CRISPR/Cas9 was used to knock out PTEN in a melanoma cell line that harbors a BRAFV600E mutation. RNA sequencing, functional kinome analysis, and drug synergy screening were employed in the context of BRAF/MEK inhibition.ResultsRNA sequencing and functional kinome analysis revealed that the loss of PTEN led to an induction of FOXD3 and an increase in expression of the FOXD3 target gene, ERBB3/HER3. Inhibition of BRAF and MEK1/2 in PTEN null, BRAFV600E cells dramatically induced the expression of ERBB3/HER3 relative to wild-type cells. A synergy screen of epigenetic modifiers and kinase inhibitors in combination with BRAFi/MEKi revealed that the pan ERBB/HER inhibitor, neratinib, could reverse the resistance observed in PTEN null, BRAFV600E cells.ConclusionsThe findings indicate that PTEN null BRAFV600E melanoma exhibits increased reliance on ERBB/HER signaling when treated with clinically approved BRAFi/MEKi combinations. Future studies are warranted to test neratinib reversal of BRAFi/MEKi resistance in patient melanomas expressing ERBB3/HER3 in combination with its dimerization partner ERBB2/HER2.
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.