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

Journals

Article Types

Countries / Regions

Search Results (157)

Search Parameters:
Keywords = druggable mutations

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
24 pages, 7124 KiB  
Article
In Silico Discovery of a Novel Potential Allosteric PI3Kα Inhibitor Incorporating 3-(2-Chloro-5-fluorophenyl)isoindolin-1-one to Target Head and Neck Squamous Cell Carcinoma
by Wenqing Jia and Xianchao Cheng
Biology 2025, 14(7), 896; https://doi.org/10.3390/biology14070896 - 21 Jul 2025
Viewed by 243
Abstract
Phosphatidylinositol 3-kinase alpha (PI3Kα) is frequently mutated in head and neck squamous cell carcinoma (HNSCC), leading to the constitutive activation of the PI3K/Akt pathway, which promotes tumor cell proliferation, survival, and metastasis. PI3Kα allosteric inhibitors demonstrate therapeutic potential as both monotherapy and combination [...] Read more.
Phosphatidylinositol 3-kinase alpha (PI3Kα) is frequently mutated in head and neck squamous cell carcinoma (HNSCC), leading to the constitutive activation of the PI3K/Akt pathway, which promotes tumor cell proliferation, survival, and metastasis. PI3Kα allosteric inhibitors demonstrate therapeutic potential as both monotherapy and combination therapy, particularly in patients with PIK3CA mutations or resistance to immunotherapy, through the precise targeting of mutant PI3Kα. Compared to ATP-competitive PI3Kα inhibitors such as Alpelisib, the allosteric inhibitor RLY-2608 exhibits enhanced selectivity for mutant PI3Kα while minimizing the inhibition of wild-type PI3Kα, thereby reducing side effects such as hyperglycemia. To date, no allosteric PI3Kα inhibitors have been approved for clinical use. To develop novel PI3Kα inhibitors with improved safety and efficacy, we employed a scaffold hopping approach to structurally modify RLY-2608 and constructed a compound library. Based on the structural information of the PI3Kα allosteric site, we conducted the systematic virtual screening of 11,550 molecules from databases to identify lead compounds. Through integrated approaches, including molecular docking studies, target validation, druggability evaluation, molecular dynamics simulations, and metabolic pathway and metabolite analyses, we successfully identified a promising novel allosteric PI3Kα inhibitor, H-18 (3-(2-chloro-5-fluorophenyl)isoindolin-1-one). H-18 has not been previously reported as a PI3Kα inhibitor, and provides an excellent foundation for subsequent lead optimization, offering a significant starting point for the development of more potent PI3Kα allosteric inhibitors. Full article
(This article belongs to the Special Issue Protein Kinases: Key Players in Carcinogenesis)
Show Figures

Figure 1

28 pages, 1744 KiB  
Review
HER2 in Non-Small Cell Lung Cancer (NSCLC): Evolution of the Therapeutic Landscape and Emerging Drugs—A Long Way to the Top
by Pamela Trillo Aliaga, Gianluca Spitaleri, Ilaria Attili, Carla Corvaja, Elena Battaiotto, Panagiotis Agisilaos Angelopoulos, Ester Del Signore, Antonio Passaro and Filippo de Marinis
Molecules 2025, 30(12), 2645; https://doi.org/10.3390/molecules30122645 - 18 Jun 2025
Viewed by 1362
Abstract
Non-small-cell lung cancer (NSCLC) can harbour different HER2 alterations: HER2 protein overexpression (2–35%), HER2 gene amplification (2–20%), and gene mutations (1–4%). The discovery of the HER2 gene in the 1980s raised great expectations for the treatment of several tumours. However, it was only [...] Read more.
Non-small-cell lung cancer (NSCLC) can harbour different HER2 alterations: HER2 protein overexpression (2–35%), HER2 gene amplification (2–20%), and gene mutations (1–4%). The discovery of the HER2 gene in the 1980s raised great expectations for the treatment of several tumours. However, it was only in 2004 that HER2 mutations were identified, and they currently represent a key druggable target in NSCLC. Despite numerous strengths, there is only one FDA/EMA-approved targeted therapy, an antibody-drug conjugate (ADC) called trastuzumab deruxtecan for pretreated patients with HER2 mutant NSCLC. In the first-line treatment, the standard of care (SoC) remains chemotherapy with or without immunotherapy. In the past, pan-HER tyrosine kinase inhibitors (TKIs) were extensively studied with poor results. But, two newly developed HER2-specific TKIs with low EGFR WT inhibition (BAY2927088 and zongertinib) reported encouraging results and received the breakthrough therapy designation from the FDA. Ongoing clinical trials are investigating new agents. This review focuses on HER2 alterations. Additionally, the anti-HER2 therapies explored so far will be discussed in detail, including the following: HER2 inhibitors (pan-inhibitors and selective inhibitors), monoclonal antibodies (mAbs), and ADCs. A section of this paper is dedicated to the role of immunotherapy in HER2-altered NSCLC. The last section of this paper focuses on the drugs under development and their challenges. Full article
(This article belongs to the Special Issue New Insights into Kinase Inhibitors II)
Show Figures

Figure 1

13 pages, 251 KiB  
Review
Perioperative Strategies in Resectable Non-Squamous Non-Small Cell Lung Cancer with EGFR Mutations and ALK Rearrangement
by Francesco Petrella, Andrea Cara, Enrico Mario Cassina, Sara Degiovanni, Lidia Libretti, Sara Lo Torto, Emanuele Pirondini, Federico Raveglia, Francesca Spinelli, Antonio Tuoro and Stefania Rizzo
Cancers 2025, 17(11), 1844; https://doi.org/10.3390/cancers17111844 - 31 May 2025
Viewed by 721
Abstract
Lung cancer is the leading cause of cancer-related death worldwide, ranking first among men and second among women for both incidence and mortality. Surgery remains the primary treatment for early-stage, resectable non-small cell lung cancer (NSCLC), encompassing stages I and selected cases of [...] Read more.
Lung cancer is the leading cause of cancer-related death worldwide, ranking first among men and second among women for both incidence and mortality. Surgery remains the primary treatment for early-stage, resectable non-small cell lung cancer (NSCLC), encompassing stages I and selected cases of stage IIIB. For patients with stage II to III disease, as well as some stage IB tumors, neoadjuvant or adjuvant systemic therapies are recommended. It is well recognized that specific driver gene mutations play a critical role in tumor progression and aggressiveness, and patients with these genetic alterations may benefit from targeted treatment approaches. These alterations are referred to as “druggable”, “targetable”, or “actionable”, representing specific targets for personalized treatments. Tyrosine kinase inhibitors (TKIs) are now the preferred first-line treatment for patients harboring mutations in EGFR, ALK, ROS1, and BRAF. Additionally, targeted therapies exist for patients with alterations in RET, ERBB2, KRAS, MET, and NTRK, either for those who have received prior treatments or as part of ongoing clinical trials. The success of targeted therapies is reshaping treatment approaches for NSCLC with targetable driver gene alterations, both in early-stage and locally advanced settings. This review focuses on current therapeutic strategies that combine targeted therapies with surgical resection in patients with resectable non-small cell lung cancer (NSCLC) harboring actionable driver gene alterations. Full article
34 pages, 2242 KiB  
Review
Druggable Molecular Networks in BRCA1/BRCA2-Mutated Breast Cancer
by Francesca Pia Carbone, Pietro Ancona, Stefano Volinia, Anna Terrazzan and Nicoletta Bianchi
Biology 2025, 14(3), 253; https://doi.org/10.3390/biology14030253 - 2 Mar 2025
Viewed by 2809
Abstract
Mutations in the tumor suppressor genes BRCA1 and BRCA2 are associated with the triple-negative breast cancer phenotype, particularly aggressive and hard-to-treat tumors lacking estrogen, progesterone, and human epidermal growth factor receptor 2. This research aimed to understand the metabolic and genetic links behind [...] Read more.
Mutations in the tumor suppressor genes BRCA1 and BRCA2 are associated with the triple-negative breast cancer phenotype, particularly aggressive and hard-to-treat tumors lacking estrogen, progesterone, and human epidermal growth factor receptor 2. This research aimed to understand the metabolic and genetic links behind BRCA1 and BRCA2 mutations and investigate their relationship with effective therapies. Using the Cytoscape software, two networks were generated through a bibliographic analysis of articles retrieved from the PubMed-NCBI database. We identified 98 genes deregulated by BRCA mutations, and 24 were modulated by therapies. In particular, BIRC5, SIRT1, MYC, EZH2, and CSN2 are influenced by BRCA1, while BCL2, BAX, and BRIP1 are influenced by BRCA2 mutation. Moreover, the study evaluated the efficacy of several promising therapies, targeting only BRCA1/BRCA2-mutated cells. In this context, CDDO-Imidazolide was shown to increase ROS levels and induce DNA damage. Similarly, resveratrol decreased the expression of the anti-apoptotic gene BIRC5 while it increased SIRT1 both in vitro and in vivo. Other specific drugs were found to induce apoptosis selectively in BRCA-mutated cells or block cell growth when the mutation occurs, i.e., 3-deazaneplanocin A, genistein or daidzein, and PARP inhibitors. Finally, over-representation analysis on the genes highlights ferroptosis and proteoglycan pathways as potential drug targets for more effective treatments. Full article
(This article belongs to the Special Issue Advances in Biological Breast Cancer Research)
Show Figures

Graphical abstract

15 pages, 1580 KiB  
Article
Conventional PCR Versus Next Generation Sequencing for Diagnosis of FLT3, IDH and NPM1 Mutations in Acute Myeloid Leukemia: Results of the PETHEMA PCR-LMA Study
by Blanca Boluda, Rebeca Rodriguez-Veiga, Claudia Sargas, Rosa Ayala, María J. Larráyoz, María Carmen Chillón, Elena Soria-Saldise, Cristina Bilbao, Esther Prados de la Torre, Irene Navarro, David Martinez-Cuadron, Cristina Gil, Teresa Bernal, Juan Bergua, Lorenzo Algarra, Mar Tormo, Pilar Martínez-Sanchez, Estrella Carrillo-Cruz, Josefina Serrano, Juan M. Alonso-Domínguez, Raimundo García, Maria Luz Amigo, Pilar Herrera-Puente, María J. Sayas, Esperanza Lavilla-Rubira, María José García-Pérez, Julia Morán, Esther Pérez-Santaolalla, Natalia Alonso-Vence, Ana Oliva, Juan Antonio López, Manuel Barrios, María García-Fortes, María Teresa Olave, Jorge Labrador, Joaquín Martínez-López, María J. Calasanz, Ramón García-Sanz, José A. Pérez-Simón, María T. Gómez-Casares, Joaquín Sánchez-Garcia, Yolanda Mendizabal, Eva Barragán and Pau Montesinosadd Show full author list remove Hide full author list
Cancers 2025, 17(5), 854; https://doi.org/10.3390/cancers17050854 - 1 Mar 2025
Viewed by 1322
Abstract
Background/Objectives: This PETHEMA PCR-LMA study aimed to evaluate whether mutations detected by NGS (VAF cut-off of ≥5%) correlate with NPM1, FLT3-ITD, FLT3-TKD, IDH1, and IDH2 mutations detected using conventional PCR (analytical sensitivity 3%) in a nationwide network of seven reference laboratories. Methods: Between [...] Read more.
Background/Objectives: This PETHEMA PCR-LMA study aimed to evaluate whether mutations detected by NGS (VAF cut-off of ≥5%) correlate with NPM1, FLT3-ITD, FLT3-TKD, IDH1, and IDH2 mutations detected using conventional PCR (analytical sensitivity 3%) in a nationwide network of seven reference laboratories. Methods: Between 2019 and 2021, 1685 adult AML patients with at least one centralized sample (NGS or PCR) at primary diagnosis or relapse/refractory episode were included. Results: During this period, 1288 paired NGS/PCR samples (1094 at diagnosis, 103 at relapse and 88 at refractoriness) were analyzed. Considering PCR the gold-standard, for NPM1 NGS sensitivity was 98.5% and specificity 98.9%, for FLT3-ITD 73.8% and 99.6%, for FLT3-TKD 84.5% and 99.3%, for IDH1 98.7% and 98.7%, and for IDH2 99.1% and 97.7%, respectively. Overall concordance rate of positive results between NGS (and PCR was 95% (262/276) for NPM1, 72% (149/206) for FLT3-ITD, 74% (49/66) for FLT3-TKD, 87% (77/89) for IDH1 and 84% (107/127) for IDH2. Overall, median days from sample reception until report were 7 for PCR and 28 for NGS. Conclusions: This study shows high concordance between NPM1 and IDH results using PCR and NGS. However, sensible important discrepancies are observed for FLT3 mutations. In our context, rapid screening for these druggable mutations should be performed by conventional PCR. Full article
(This article belongs to the Special Issue New Approaches in Leukemia)
Show Figures

Figure 1

28 pages, 37943 KiB  
Article
RAC1-Amplified and RAC1-A159V Hotspot-Mutated Head and Neck Cancer Sensitive to the Rac Inhibitor EHop-016 In Vivo: A Proof-of-Concept Study
by Helen Hoi Yin Chan, Hoi-Lam Ngan, Yuen-Keng Ng, Chun-Ho Law, Peony Hiu Yan Poon, Ray Wai Wa Chan, Kwok-Fai Lau, Wenying Piao, Hui Li, Lan Wang, Jason Ying Kuen Chan, Yu-Xiong Su, Thomas Chun Kit Yeung, Eileen Wong, Angela Wing Tung Li, Krista Roberta Verhoeft, Yuchen Liu, Yukai He, Stephen Kwok-Wing Tsui, Gordon B. Mills and Vivian Wai Yan Luiadd Show full author list remove Hide full author list
Cancers 2025, 17(3), 361; https://doi.org/10.3390/cancers17030361 - 23 Jan 2025
Cited by 1 | Viewed by 1481
Abstract
Objective: RAC1 aberrations in head and neck squamous cell carcinoma (HNSCC) remain clinically inactionable today. Methods: Here, we investigated the clinical significance and potential druggability of RAC1 genomic aberrations in HNSCC. Results: Notably, HPV(−)HNSCC patients bearing the unique HNSCC-prevalent RAC1-A159V hotspot [...] Read more.
Objective: RAC1 aberrations in head and neck squamous cell carcinoma (HNSCC) remain clinically inactionable today. Methods: Here, we investigated the clinical significance and potential druggability of RAC1 genomic aberrations in HNSCC. Results: Notably, HPV(−)HNSCC patients bearing the unique HNSCC-prevalent RAC1-A159V hotspot mutation, P29S hotspot and G-box domain mutations, and RAC1 copy number increases all displayed dismal overall survival (TCGA-HNSCC). Here, we demonstrated that all five HNSCC patient-relevant RAC1 aberrations tested (A159V and P29S hotspot mutations, K116N, G15S, and N39S) could significantly drive HNSCC tumoroid growth and/invasion, with A159V, P29S, and K116N mutants being the most potent drivers. Interestingly, transcriptomics analyses revealed that RAC1 mutations and copy increase could both drive PI3K pathway activation, with the A159V mutant associated with the prominent intra-tumoral upregulation of phospho-RPS6(Ser235/236) in patient tumors. Importantly, proof-of-principle Rac targeting with EHop-016 resulted in remarkable antitumor activity in vivo against RAC1-A159V-mutated and RAC1-amplified HNSCC patient-derived xenografts (PDXs) and/engineered models. Lastly, melanoma and endometrial xenograft models bearing endogenous RAC1-amplification and RAC1-A159V mutation were also sensitive to EHop-016 targeting. Conclusions: In principle, RAC1 genomic aberrations in HNSCC can be potentially harnessed for precision drugging. Full article
Show Figures

Figure 1

21 pages, 6841 KiB  
Article
Marine Origin vs. Synthesized Compounds: In Silico Screening for a Potential Drug Against SARS-CoV-2
by Amar Osmanović, Mirsada Salihović, Elma Veljović, Lamija Hindija, Mirha Pazalja, Maja Malenica, Aida Selmanagić and Selma Špirtović-Halilović
Sci. Pharm. 2025, 93(1), 2; https://doi.org/10.3390/scipharm93010002 - 26 Dec 2024
Cited by 1 | Viewed by 1656
Abstract
Although COVID-19 is not a pandemic anymore, the virus frequently mutates, resulting in new strains and presenting global public health challenges. The lack of oral antiviral drugs makes it difficult to treat him, which makes the creation of broadly acting antivirals necessary to [...] Read more.
Although COVID-19 is not a pandemic anymore, the virus frequently mutates, resulting in new strains and presenting global public health challenges. The lack of oral antiviral drugs makes it difficult to treat him, which makes the creation of broadly acting antivirals necessary to fight current and next epidemics of viruses. Using the molecular docking approach, 118 compounds derived from marine organisms and 92 previously synthesized compounds were screened to assess their binding affinity for the main protease and papain-like protease enzymes of SARS-CoV-2. The best candidates from the xanthene, benzoxazole, and coumarin classes were identified. Marine-derived compounds showed slightly better potential as enzyme inhibitors, though the binding affinities of synthesized compounds were similar, with the best candidates displaying affinity values between 0.2 and 0.4 mM. Xanthenes, among both marine origin and synthesized compounds, emerged as the most promising scaffolds for further research as inhibitors. The papain-like protease was found to be more druggable than the main protease. Additionally, all top candidates met the criteria for various drug-likeness properties, indicating good oral bioavailability and low risk of adverse effects. This research provides valuable insights into the comparative affinities of marine origin and synthesized compounds from the xanthene, coumarin, and benzoxazole classes, highlighting promising candidates for further in vitro and in vivo studies. Full article
Show Figures

Figure 1

17 pages, 4611 KiB  
Article
Identifying Allosteric Small-Molecule Binding Sites of Inactive NS2B-NS3 Proteases of Pathogenic Flaviviridae
by Hovakim Grabski, Siranuysh Grabska and Ruben Abagyan
Viruses 2025, 17(1), 6; https://doi.org/10.3390/v17010006 - 24 Dec 2024
Viewed by 1476
Abstract
Dengue, West Nile, Zika, Yellow fever, and Japanese encephalitis viruses persist as significant global health threats. The development of new therapeutic strategies based on inhibiting essential viral enzymes or viral–host protein interactions is problematic due to the fast mutation rate and rapid emergence [...] Read more.
Dengue, West Nile, Zika, Yellow fever, and Japanese encephalitis viruses persist as significant global health threats. The development of new therapeutic strategies based on inhibiting essential viral enzymes or viral–host protein interactions is problematic due to the fast mutation rate and rapid emergence of drug resistance. This study focuses on the NS2B-NS3 protease as a promising target for antiviral drug development. Promising allosteric binding sites were identified in two conformationally distinct inactive states and characterized for five flaviviruses and four Dengue virus subtypes. Their shapes, druggability, inter-viral similarity, sequence variation, and susceptibility to drug-resistant mutations have been studied. Two identified allosteric inactive state pockets appear to be feasible alternatives to a larger closed pocket near the active site, and they can be targeted with specific drug-like small-molecule inhibitors. Virus-specific sequence and structure implications and the feasibility of multi-viral inhibitors are discussed. Full article
(This article belongs to the Special Issue Recent Advances in Anti-HCV, Anti-HBV and Anti-flavivirus Agents)
Show Figures

Figure 1

28 pages, 5658 KiB  
Review
Mechanistic Insights into the Mutational Landscape of the Main Protease/3CLPro and Its Impact on Long-Term COVID-19/SARS-CoV-2 Management
by Aganze Gloire-Aimé Mushebenge, Samuel Chima Ugbaja, Nonjabulo Ntombikhona Magwaza, Nonkululeko Avril Mbatha, Tambwe Willy Muzumbukilwa, Mukanda Gedeon Kadima, Fave Yohanna Tata, Mthokosizi Bongani Nxumalo, Riziki Ghislain Manimani, Ntabaza Ndage, Bakari Salvius Amuri, Kahumba Byanga, Manimbulu Nlooto, Rene B. Khan and Hezekiel M. Kumalo
Future Pharmacol. 2024, 4(4), 825-852; https://doi.org/10.3390/futurepharmacol4040044 - 28 Nov 2024
Viewed by 2720
Abstract
The main proteinase (Mpro), or 3CLpro, is a critical enzyme in the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) lifecycle and is responsible for breaking down and releasing vital functional viral proteins crucial for virus development and transmission. As a catalytically active dimer, [...] Read more.
The main proteinase (Mpro), or 3CLpro, is a critical enzyme in the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) lifecycle and is responsible for breaking down and releasing vital functional viral proteins crucial for virus development and transmission. As a catalytically active dimer, its dimerization interface has become an attractive target for antiviral drug development. Recent research has extensively investigated the enzymatic activity of Mpro, focusing on its role in regulating the coronavirus replication complex and its significance in virus maturation and infectivity. Computational investigations have identified four druggable pockets, suggesting potential allosteric sites beyond the substrate-binding region. Empirical validation through site-directed alanine mutagenesis has targeted residues in both the active and allosteric regions and corroborated these predictions. Structural studies of drug target proteins can inform therapeutic approaches, with metadynamics simulations shedding light on the role of H163 in regulating Mpro function and providing insights into its dynamic equilibrium to the wild-type enzyme. Despite the efficacy of vaccines and drugs in mitigating SARS-CoV-2 spread, its ongoing viral evolution, selective pressures, and continued transmission pose challenges, potentially leading to resistant mutations. Phylogenetic analyses have indicated the existence of several resistant variations predating drug introduction to the human population, emphasizing the likelihood of drug spread. Hydrogen/deuterium-exchange mass spectrometry reveals the structural influence of the mutation. At the same time, clinical trials on 3CLPro inhibitors underscore the clinical significance of reduced enzymatic activity and offer avenues for future therapeutic exploration. Understanding the implications of 3CLPro mutations holds promise for shaping forthcoming therapeutic strategies against COVID-19. This review delves into factors influencing mutation rates and identifies areas warranting further investigation, providing a comprehensive overview of Mpro mutations, categorization, and terminology. Moreover, we examine their associations with clinical outcomes, illness severity, unresolved issues, and future research prospects, including their impact on vaccine efficacy and potential therapeutic targeting. Full article
Show Figures

Figure 1

12 pages, 663 KiB  
Review
Clinical Advances and Challenges in Targeting KRAS Mutations in Non-Small Cell Lung Cancer
by Simone E. Dekker and Lei Deng
Cancers 2024, 16(22), 3885; https://doi.org/10.3390/cancers16223885 - 20 Nov 2024
Cited by 2 | Viewed by 2516 | Correction
Abstract
KRAS mutation is one of the most common oncogenic drivers in non-small cell lung cancer. Since its discovery about four decades ago, drug development targeting KRAS has been met with countless failures. Recently, KRAS G12C, a subvariant of KRAS, became the first druggable [...] Read more.
KRAS mutation is one of the most common oncogenic drivers in non-small cell lung cancer. Since its discovery about four decades ago, drug development targeting KRAS has been met with countless failures. Recently, KRAS G12C, a subvariant of KRAS, became the first druggable KRAS mutation. The efficacy of the first-generation KRAS inhibitor is modest, but with scientific advancement, KRAS G12C inhibitors with higher potency are on the horizon. Additionally, novel therapeutic approaches targeting other KRAS subvariants are also being explored in clinical trials with encouraging early data. We will review the clinical advances and challenges for patients with KRAS-mutated non-small cell lung cancer, with a focus on small molecule inhibitors. Full article
Show Figures

Figure 1

14 pages, 913 KiB  
Review
Decoding Acute Myeloid Leukemia: A Clinician’s Guide to Functional Profiling
by Prasad Iyer, Shaista Shabbir Jasdanwala, Yuhan Wang, Karanpreet Bhatia and Shruti Bhatt
Diagnostics 2024, 14(22), 2560; https://doi.org/10.3390/diagnostics14222560 - 14 Nov 2024
Cited by 2 | Viewed by 1915
Abstract
Acute myeloid leukemia (AML) is a complex clonal disorder characterized by clinical, genetic, metabolomic, and epigenetic heterogeneity resulting in the uncontrolled proliferation of aberrant blood-forming precursor cells. Despite advancements in the understanding of the genetic, metabolic, and epigenetic landscape of AML, it remains [...] Read more.
Acute myeloid leukemia (AML) is a complex clonal disorder characterized by clinical, genetic, metabolomic, and epigenetic heterogeneity resulting in the uncontrolled proliferation of aberrant blood-forming precursor cells. Despite advancements in the understanding of the genetic, metabolic, and epigenetic landscape of AML, it remains a significant therapeutic challenge. Functional profiling techniques, such as BH3 profiling (BP), gene expression profiling (GEP), proteomics, metabolomics, drug sensitivity/resistance testing (DSRT), CRISPR/Cas9, and RNAi screens offer valuable insights into the functional behavior of leukemia cells. BP evaluates the mitochondrial response to pro-apoptotic BH3 peptides, determining a cell’s apoptotic threshold and its reliance on specific anti-apoptotic proteins. This knowledge can pinpoint vulnerabilities in the mitochondria-mediated apoptotic pathway in leukemia cells, potentially informing treatment strategies and predicting therapeutic responses. GEP, particularly RNA sequencing, evaluates the transcriptomic landscape and identifies gene expression alterations specific to AML subtypes. Proteomics and metabolomics, utilizing mass spectrometry and nuclear magnetic resonance (NMR), provide a detailed view of the active proteins and metabolic pathways in leukemia cells. DSRT involves exposing leukemia cells to a panel of chemotherapeutic and targeted agents to assess their sensitivity or resistance profiles and potentially guide personalized treatment strategies. CRISPR/Cas9 and RNAi screens enable systematic disruption of genes to ascertain their roles in leukemia cell survival and proliferation. These techniques facilitate precise disease subtyping, uncover novel biomarkers and therapeutic targets, and provide a deeper understanding of drug-resistance mechanisms. Recent studies utilizing functional profiling have identified specific mutations and gene signatures associated with aggressive AML subtypes, aberrant signaling pathways, and potential opportunities for drug repurposing. The integration of multi-omics approaches, advances in single-cell sequencing, and artificial intelligence is expected to refine the precision of functional profiling and ultimately improve patient outcomes in AML. This review highlights the diverse landscape of functional profiling methods and emphasizes their respective advantages and limitations. It highlights select successes in how these methods have further advanced our understanding of AML biology, identifies druggable targets that have improved outcomes, delineates challenges associated with these techniques, and provides a prospective view of the future where these techniques are likely to be increasingly incorporated into the routine care of patients with AML. Full article
(This article belongs to the Section Clinical Laboratory Medicine)
Show Figures

Figure 1

18 pages, 5930 KiB  
Article
Use of 3′ Rapid Amplification of cDNA Ends (3′ RACE)-Based Targeted RNA Sequencing for Profiling of Druggable Genetic Alterations in Urothelial Carcinomas
by Natalia V. Mitiushkina, Vladislav I. Tiurin, Aleksandra A. Anuskina, Natalia A. Bordovskaya, Ekaterina A. Nalivalkina, Darya M. Terina, Mariya V. Berkut, Anna D. Shestakova, Maria V. Syomina, Ekaterina Sh. Kuligina, Alexandr V. Togo and Evgeny N. Imyanitov
Int. J. Mol. Sci. 2024, 25(22), 12126; https://doi.org/10.3390/ijms252212126 - 12 Nov 2024
Cited by 1 | Viewed by 1647
Abstract
Targeted treatment of advanced or metastatic urothelial carcinomas (UCs) requires the identification of druggable mutations. This study describes the development of a 3′ Rapid Amplification of cDNA Ends (3′ RACE)-based targeted RNA sequencing panel which accounts for the status of all genes relevant [...] Read more.
Targeted treatment of advanced or metastatic urothelial carcinomas (UCs) requires the identification of druggable mutations. This study describes the development of a 3′ Rapid Amplification of cDNA Ends (3′ RACE)-based targeted RNA sequencing panel which accounts for the status of all genes relevant to UC treatment, namely, FGFR1-4, KRAS, NRAS, BRAF, ERBB2 (HER2), CD274 (PD-L1) and PIK3CA. FGFR2/3-activating point mutations or fusions were found in 54/233 (23.2%) tumors. FGFR3 rearrangements were identified in 11 patients, with eight of them being undetectable by commonly used PCR kits. In addition, one tumor contained a high-copy FGFR2 gene amplification accompanied by strong overexpression of the gene. Mutations in RAS/RAF genes were present in 30/233 (12.9%) UCs and were mutually exclusive with alterations affecting FGFR2/3 genes. On the contrary, activating events in the HER2 oncogene (point mutations and overexpression), as well as PIK3CA mutations, which were relatively common, occurred with similar frequencies in RAS/RAF- or FGFR2/3-positive vs. negative samples. High PD-L1 mRNA expression was associated with advanced disease stage and was not observed in tumors with increased HER2 mRNA expression or in UCs with evidence for FGFR2/3 activation. Three of the studied carcinomas had high-level microsatellite instability (MSI). Overall, more than half of the UCs had potentially druggable genetic alterations. The proposed NGS panel permits comprehensive and cost-efficient analysis of UC-specific molecular targets and may be considered in clinical routine. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

17 pages, 13647 KiB  
Article
Single-Nuclei Transcriptome Profiling Reveals Intra-Tumoral Heterogeneity and Characterizes Tumor Microenvironment Architecture in a Murine Melanoma Model
by Sushant Parab, Valery Sarlo, Sonia Capellero, Luca Palmiotto, Alice Bartolini, Daniela Cantarella, Marcello Turi, Annamaria Gullà, Elena Grassi, Chiara Lazzari, Marco Rubatto, Vanesa Gregorc, Fabrizio Carnevale-Schianca, Martina Olivero, Federico Bussolino and Valentina Comunanza
Int. J. Mol. Sci. 2024, 25(20), 11228; https://doi.org/10.3390/ijms252011228 - 18 Oct 2024
Viewed by 1723
Abstract
Malignant melanoma is an aggressive cancer, with a high risk of metastasis and mortality rates, characterized by cancer cell heterogeneity and complex tumor microenvironment (TME). Single cell biology is an ideal and powerful tool to address these features at a molecular level. However, [...] Read more.
Malignant melanoma is an aggressive cancer, with a high risk of metastasis and mortality rates, characterized by cancer cell heterogeneity and complex tumor microenvironment (TME). Single cell biology is an ideal and powerful tool to address these features at a molecular level. However, this approach requires enzymatic cell dissociation that can influence cellular coverage. By contrast, single nucleus RNA sequencing (snRNA-seq) has substantial advantages including compatibility with frozen samples and the elimination of a dissociation-induced, transcriptional stress response. To better profile and understand the functional diversity of different cellular components in melanoma progression, we performed snRNA-seq of 16,839 nuclei obtained from tumor samples along the growth of murine syngeneic melanoma model carrying a BRAFV600E mutation and collected 9 days or 23 days after subcutaneous cell injection. We defined 11 different subtypes of functional cell clusters among malignant cells and 5 different subsets of myeloid cells that display distinct global transcriptional program and different enrichment in early or advanced stage of tumor growth, confirming that this approach was useful to accurately identify intratumor heterogeneity and dynamics during tumor evolution. The current study offers a deep insight into the biology of melanoma highlighting TME reprogramming through tumor initiation and progression, underlying further discovery of new TME biomarkers which may be potentially druggable. Full article
Show Figures

Figure 1

11 pages, 754 KiB  
Article
Multigene Panel Next-Generation Sequencing Techniques in the Management of Patients with Metastatic Colorectal Carcinoma: The Way Forward for Personalized Treatment? A Single-Center Experience
by Laura Matteucci, Francesco Giulio Sullo, Chiara Gallio, Luca Esposito, Margherita Muratore, Ilario Giovanni Rapposelli, Daniele Calistri, Elisabetta Petracci, Claudia Rengucci, Laura Capelli, Elisa Chiadini, Paola Ulivi, Alessandro Passardi and Alessandro Bittoni
Int. J. Mol. Sci. 2024, 25(20), 11071; https://doi.org/10.3390/ijms252011071 - 15 Oct 2024
Cited by 1 | Viewed by 1249
Abstract
The efficacy and cost-effectiveness of Multigene Panel Next-Generation Sequencing (NGS) in directing patients towards genomically matched therapies remain uncertain. This study investigated metastatic colorectal cancer (mCRC) patients who underwent NGS analysis on formalin-fixed paraffin-embedded tumor samples. Data from 179 patients were analyzed, revealing [...] Read more.
The efficacy and cost-effectiveness of Multigene Panel Next-Generation Sequencing (NGS) in directing patients towards genomically matched therapies remain uncertain. This study investigated metastatic colorectal cancer (mCRC) patients who underwent NGS analysis on formalin-fixed paraffin-embedded tumor samples. Data from 179 patients were analyzed, revealing no mutations in 39 patients (21.8%), one mutation in 83 patients (46.4%), and two or more mutations in 57 patients (31.8%). KRAS mutations were found in 87 patients (48.6%), including KRAS G12C mutations in 5 patients (2.8%), PIK3CA mutations in 40 patients (22.4%), and BRAF mutations in 26 patients (14.5%). Less common mutations were identified: ERBB2 in five patients (2.8%) and SMO in four patients (2.2%). Additionally, MAP2K1, CTNNB1, and MYC were mutated in three patients (2.4%). Two mutations (1.1%) were observed in ERBB3, RAF1, MTOR, JAK1, and FGFR2. No significant survival differences were observed based on number of mutations. In total, 40% of patients had druggable molecular alterations, but only 1.1% received genomically guided treatment, suggesting limited application in standard practice. Despite this, expanded gene panel testing can identify actionable mutations, aiding personalized treatment strategies in metastatic CRC, although current eligibility for biomarker-guided trials remains limited. Full article
Show Figures

Figure 1

15 pages, 938 KiB  
Review
A New Frontier in Cystic Fibrosis Pathophysiology: How and When Clock Genes Can Affect the Inflammatory/Immune Response in a Genetic Disease Model
by Annalucia Carbone, Pamela Vitullo, Sante Di Gioia, Stefano Castellani and Massimo Conese
Curr. Issues Mol. Biol. 2024, 46(9), 10396-10410; https://doi.org/10.3390/cimb46090618 - 18 Sep 2024
Viewed by 3384
Abstract
Cystic fibrosis (CF) is a monogenic syndrome caused by variants in the CF Transmembrane Conductance Regulator (CFTR) gene, affecting various organ and systems, in particular the lung, pancreas, sweat glands, liver, gastrointestinal tract, vas deferens, and vascular system. While for some [...] Read more.
Cystic fibrosis (CF) is a monogenic syndrome caused by variants in the CF Transmembrane Conductance Regulator (CFTR) gene, affecting various organ and systems, in particular the lung, pancreas, sweat glands, liver, gastrointestinal tract, vas deferens, and vascular system. While for some organs, e.g., the pancreas, a strict genotype-phenotype occurs, others, such as the lung, display a different pathophysiologic outcome in the presence of the same mutational asset, arguing for genetic and environmental modifiers influencing severity and clinical trajectory. CFTR variants trigger a pathophysiological cascade of events responsible for chronic inflammatory responses, many aspects of which, especially related to immunity, are not ascertained yet. Although clock genes expression and function are known modulators of the innate and adaptive immunity, their involvement in CF has been only observed in relation to sleep abnormalities. The aim of this review is to present current evidence on the clock genes role in immune-inflammatory responses at the lung level. While information on this topic is known in other chronic airway diseases (chronic obstructive pulmonary disease and asthma), CF lung disease (CFLD) is lacking in this knowledge. We will present the bidirectional effect between clock genes and inflammatory factors that could possibly be implicated in the CFLD. It must be stressed that besides sleep disturbance and its mechanisms, there are not studies directly addressing the exact nature of clock genes’ involvement in inflammation and immunity in CF, pointing out the directions of new and deepened studies in this monogenic affection. Importantly, clock genes have been found to be druggable by means of genetic tools or pharmacological agents, and this could have therapeutic implications in CFLD. Full article
(This article belongs to the Special Issue Complex Molecular Mechanism of Monogenic Diseases: 3rd Edition)
Show Figures

Figure 1

Back to TopTop