Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (105)

Search Parameters:
Keywords = Menin

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 427 KiB  
Review
Therapeutic Implications of Menin Inhibitors in the Treatment of Acute Leukemia: A Critical Review
by Martina Canichella, Cristina Papayannidis, Carla Mazzone and Paolo de Fabritiis
Diseases 2025, 13(7), 227; https://doi.org/10.3390/diseases13070227 - 19 Jul 2025
Viewed by 475
Abstract
Menin inhibitors are a class of targeted agents that exemplify how a deeper understanding of leukemia pathogenesis can unify seemingly distinct genetic acute leukemia subgroups under a common therapeutic strategy. In particular, acute leukemia with NPM1 mutations (NPM1m) and KMT2A rearrangements ( [...] Read more.
Menin inhibitors are a class of targeted agents that exemplify how a deeper understanding of leukemia pathogenesis can unify seemingly distinct genetic acute leukemia subgroups under a common therapeutic strategy. In particular, acute leukemia with NPM1 mutations (NPM1m) and KMT2A rearrangements (KMT2Ar) represent the primary targets of this emerging drug class. Acute myeloid leukemia (AML) with NPM1m—which accounts for approximately 30% of AML cases and AML or acute lymphoblastic leukemia (ALL) with KMT2Ar—and is present in 5–10% of cases, shares a common pathogenetic mechanism: the aberrant activation of the MEIS1–HOXA axis. These leukemic subsets are associated with poor prognosis, particularly in the relapsed/refractory (R/R) setting. For KMT2Ar AML, the prognosis is especially dismal, with a median overall survival (OS) of 2.4 months and a complete remission (CR) rate of only 5%. In NPM1m AML, intensive chemotherapy achieves remission in approximately 80% of cases, but relapse remains a major challenge, occurring in nearly 50% of patients. Relapsed NPM1m AML is linked to a poor prognosis, with a median OS of 6.1 months (12-month OS: 30%) and a median relapse-free survival (RFS) of 5.5 months (12-month RFS: 34%). Menin inhibitors directly target the leukemogenic transcriptional program driven by HOX and MEIS1, disrupting oncogenic signaling and offering a promising therapeutic approach for these high-risk patients. This class of agents has rapidly progressed through clinical development, showing promising antileukemic activity in both treatment-naïve and R/R AML. Currently, six menin inhibitors are in clinical evaluation as monotherapy or in combination regimens: revumenib, ziftomenib, bleximenib (previously JNJ-75276617), enzomenib (previously DSP-5336), DS-1594, and BMF-219. In this review, we critically analyze the clinical development and therapeutic potential of the four most extensively studied menin inhibitors—revumenib, ziftomenib, bleximenib, and enzomenib. We discuss their efficacy, safety profiles, and potential roles within the current treatment algorithm. The continued clinical evaluation of menin inhibitors may redefine treatment paradigms for NPM1m and KMT2Ar AML and other acute leukemia with the aberrant MEIS1-HOXA axis, offering new hope for patients with limited therapeutic options. Full article
(This article belongs to the Special Issue Targeted Therapies for Acute Leukemias)
Show Figures

Figure 1

42 pages, 704 KiB  
Review
NPM1-Mutated AML: Deciphering the Molecular and Clinical Puzzle in the Era of Novel Treatment Strategies
by Michael D. Diamantidis, Maria Smaragdi Vlachou, Anastasia Katsikavela, Smaragdi Kalomoiri, Vasiliki Bartzi and Georgia Ikonomou
Cancers 2025, 17(13), 2095; https://doi.org/10.3390/cancers17132095 - 23 Jun 2025
Viewed by 1049
Abstract
The aberrant localization of the mutated nucleophosmin (NPM1) protein in the cytoplasm is the hallmark of the development of acute myeloid leukemia (AML); the gene, located in the nucleolus, codes for a protein that normally shuttles between the nucleus and the [...] Read more.
The aberrant localization of the mutated nucleophosmin (NPM1) protein in the cytoplasm is the hallmark of the development of acute myeloid leukemia (AML); the gene, located in the nucleolus, codes for a protein that normally shuttles between the nucleus and the cytoplasm of the normal hematopoietic cells. Patients harboring NPM1 mutations are diagnosed as having NPM1-mutated AMLs, which are types of leukemia with distinct clinical and laboratory characteristics. The essential diagnostics for investigating NPM1-mutated AMLs, the interactions with concomitant mutations affecting prognosis and the therapeutic interventions that the treatment of such patients requires are discussed in this review. Novel investigational agents in current clinical trials are also highlighted, along with the roles of exportin 1 (XPO1), menin-KMT2A inhibitors and immunotherapy in NPM1-mutated AMLs. This review focuses on critically evaluating the available data and aims to reveal the secrets of NPM1-mutated AMLs. Full article
(This article belongs to the Special Issue Acute Myeloid Leukemia in Adults)
Show Figures

Figure 1

17 pages, 2738 KiB  
Article
Modeling of Phase-Interpolator-Based Clock and Data Recovery for High-Speed PAM-4 Serial Interfaces
by Alessio Cortiula, Davide Menin, Andrea Bandiziol, Francesco Driussi and Pierpaolo Palestri
Electronics 2025, 14(10), 1979; https://doi.org/10.3390/electronics14101979 - 13 May 2025
Viewed by 545
Abstract
We have employed a time-domain behavioral simulator to analyze how different design options for bang-bang Clock and Data Recovery (CDR) impact the Jitter Tolerance (JTOL) performance of High-Speed Serial Interfaces (HSSIs) with PAM-4 signaling. The simulator includes the effect of Inter-Symbol Interference (ISI) [...] Read more.
We have employed a time-domain behavioral simulator to analyze how different design options for bang-bang Clock and Data Recovery (CDR) impact the Jitter Tolerance (JTOL) performance of High-Speed Serial Interfaces (HSSIs) with PAM-4 signaling. The simulator includes the effect of Inter-Symbol Interference (ISI) due to the transmission channel, various equalization schemes and a detailed description of the CDR architecture. Many design options have been investigated, with particular focus on transition filtering and on the algorithm to identify the Early/Late (E/L) information from data and edge samples after deserialization. It has been found that if majority voting is employed to derive a single set of E/L information from an array of phase detectors working on deserialized data and edges, the different filtering strategies provide the same JTOL, meaning that one can avoid transition filtering and furthermore use a single edge sampler with a zero threshold, significantly simplifying the CDR architecture. Instead, if summation of the E/L information from deserialized data and edges is performed, the decision to use one or three thresholds for the edge sampling and the choice of whether to implement transition filtering both impact JTOL; however, better performance is achieved under these conditions than when employing majority voting on the deserialized E/L signals. Full article
(This article belongs to the Section Microelectronics)
Show Figures

Figure 1

24 pages, 1770 KiB  
Review
Unraveling Venetoclax Resistance: Navigating the Future of HMA/Venetoclax-Refractory AML in the Molecular Era
by Theodora Chatzilygeroudi, Theodoros Karantanos and Vasiliki Pappa
Cancers 2025, 17(9), 1586; https://doi.org/10.3390/cancers17091586 - 7 May 2025
Cited by 1 | Viewed by 2338
Abstract
Acute myeloid leukemia (AML) has traditionally been linked to a poor prognosis, particularly in older patients who are ineligible for intensive chemotherapy. The advent of Venetoclax, a powerful oral BH3 mimetic targeting anti-apoptotic protein BCL2, has significantly advanced AML treatment. Its combination with [...] Read more.
Acute myeloid leukemia (AML) has traditionally been linked to a poor prognosis, particularly in older patients who are ineligible for intensive chemotherapy. The advent of Venetoclax, a powerful oral BH3 mimetic targeting anti-apoptotic protein BCL2, has significantly advanced AML treatment. Its combination with the hypomethylating agent azacitidine (AZA/VEN) has become a standard treatment for this group of AML patients, demonstrating a 65% overall response rate and a median overall survival of 14.7 months, compared to 22% and 8 months with azacitidine monotherapy, respectively. However, resistance and relapses remain common, representing a significant clinical challenge. Recent studies have identified molecular alterations, such as mutations in FLT3-ITD, NRAS/KRAS, TP53, and BAX, as major drivers of resistance. Additionally, other factors, including metabolic changes, anti-apoptotic protein expression, and monocytic or erythroid/megakaryocytic differentiation status, contribute to treatment failure. Clinical trials are exploring strategies to overcome venetoclax resistance, including doublet or triplet therapies targeting IDH and FLT3 mutations; novel epigenetic approaches; menin, XPO1, and MDM2 inhibitors; along with immunotherapies like monoclonal antibodies and antibody–drug conjugates. A deeper understanding of the molecular mechanisms of resistance through single-cell analysis will be crucial for developing future therapeutic strategies. Full article
(This article belongs to the Special Issue Acute Myeloid Leukemia in Adults)
Show Figures

Figure 1

32 pages, 817 KiB  
Review
An Updated Perspective of the Clinical Features and Parathyroidectomy Impact in Primary Hyperparathyroidism Amid Multiple Endocrine Neoplasia Type 1 (MEN1): Focus on Bone Health
by Ana-Maria Gheorghe, Mihaela Stanciu, Ioana Codruta Lebada, Claudiu Nistor and Mara Carsote
J. Clin. Med. 2025, 14(9), 3113; https://doi.org/10.3390/jcm14093113 - 30 Apr 2025
Viewed by 910
Abstract
Background: Multiple endocrine neoplasia type 1 (MEN1)-related primary hyperparathyroidism (MPHPT) belongs to genetic PHPT that accounts for 10% of all PHPT cases, being considered the most frequent hereditary PHPT (less than 5% of all PHPT). Objective: We aimed to provide an [...] Read more.
Background: Multiple endocrine neoplasia type 1 (MEN1)-related primary hyperparathyroidism (MPHPT) belongs to genetic PHPT that accounts for 10% of all PHPT cases, being considered the most frequent hereditary PHPT (less than 5% of all PHPT). Objective: We aimed to provide an updated clinical perspective with a double purpose: to highlight the clinical features in MPHPT, particularly, the bone health assessment, as well as the parathyroidectomy (PTx) impact. Methods: A comprehensive review of the latest 5-year, English-published, PubMed-accessed original studies. Results: The sample-based analysis (n = 17 studies) enrolled 2426 subjects (1720 with MPHPT). The study design was retrospective, except for one prospective and one case–control study. The maximum number of patients per study was of 517. Female predominance (an overall female-to-male ratio of 1.139) was confirmed (except for three studies). Age at MPHPT diagnosis (mean/median per study): 28.7 to 43.1 years; age at PTx: 32 to 43.5 years. Asymptomatic PHPT was reported in 38.3% to 67% of MPHPT. Mean total calcium varied between 1.31 and 2.88 mmol/L and highest PTH was of 317.2 pg/mL. Two studies reported similar PTH and calcaemic levels in MPHPT vs. sporadic PHPT, while another found higher values in MPHPT. Symptomatic vs. asymptomatic patients with MPHPT had similar PTH and serum calcium levels (n = 1). Osteoporosis (n = 8, N = 723 with MPHPT) was reported in 10% to 55.5% of cases, osteopenia in 5.88% to 43.9% (per study); overall fracture rate was 10% (of note, one study showed 0%). Lower bone mineral density (BMD) at DXA (n = 4) in MPHPT vs. sporadic PHPT/controls was found by some studies (n = 3, and only a single study provided third distal radius DXA-BMD assessment), but not all (n = 1). Post-PTx DXA (n = 3, N = 190 with MPHPT) showed a BMD increase (e.g., +8.5% for lumbar spine, +2.1% for total hip, +4.3% for femoral neck BMD); however, post-operatory, BMD remains lower than controls. Trabecular bone score (TBS) analysis (n = 2, N = 142 with MPHPT vs. 397 with sporadic PHPT) showed a higher prevalence of reduced TBS (n = 1) or similar (n = 1). PTx analysis in MPHPT (n = 14): rate of subtotal PTx of 39% to 66.7% (per study) or less than subtotal PTx of 46.9% (n = 1). Post-PTx complications: persistent PHPT (5.6% to 25%), recurrent PHPT (16.87% to 30%, with the highest re-operation rate of 71% in one cohort); hypoparathyroidism (12.4% to 41.7%). Genetic analysis pointed out a higher risk of post-PTx recurrence in exon 10 MEN1 pathogenic variant. Post-PTx histological exam showed a multi-glandular disease in 40% to 52.1% of MPHPT, and a parathyroid carcinoma prevalence of 1%. Conclusions: MPHPT remains a challenging ailment amid a multi-layered genetic syndrome. Current data showed a lower age at MPHPT diagnosis and surgery than found in general population, and a rate of female predominance that is lower than seen in sporadic PHPT cases, but higher than known, for instance, in MEN2. The bone involvement showed heterogeneous results, more consistent for a lower BMD, but not necessarily for a lower TBS vs. controls. PTx involves a rather high rate of recurrence, persistence and redo surgery. About one out of ten patients with MPHPT might have a prevalent fracture and PTx improves the overall bone health, but seems not to restore it to the general population level, despite the young age of the subjects. This suggests that non-parathyroid components and potentially menin protein displays negative bone effects in MEN1. Full article
(This article belongs to the Special Issue Neuroendocrine Tumors: Etiology, Diagnosis, and Therapy—2nd Edition)
Show Figures

Figure 1

27 pages, 2417 KiB  
Review
HIF-1α Pathway in COVID-19: A Scoping Review of Its Modulation and Related Treatments
by Felipe Paes Gomes da Silva, Rafael Matte, David Batista Wiedmer, Arthur Paes Gomes da Silva, Rafaela Makiak Menin, Fernanda Bressianini Barbosa, Thainá Aymê Mocelin Meneguzzi, Sabrina Barancelli Pereira, Amanda Terres Fausto, Larissa Klug, Bruna Pinheiro Melim and Claudio Jose Beltrão
Int. J. Mol. Sci. 2025, 26(9), 4202; https://doi.org/10.3390/ijms26094202 - 28 Apr 2025
Viewed by 1309
Abstract
The COVID-19 pandemic, driven by SARS-CoV-2, has led to a global health crisis, highlighting the virus’s unique molecular mechanisms that distinguish it from other respiratory pathogens. It is known that the Hypoxia-Inducible Factor 1α (HIF-1α) activates a complex network of intracellular signaling pathways [...] Read more.
The COVID-19 pandemic, driven by SARS-CoV-2, has led to a global health crisis, highlighting the virus’s unique molecular mechanisms that distinguish it from other respiratory pathogens. It is known that the Hypoxia-Inducible Factor 1α (HIF-1α) activates a complex network of intracellular signaling pathways regulating cellular energy metabolism, angiogenesis, and cell survival, contributing to the wide range of clinical manifestations of COVID-19, including Post-Acute COVID-19 Syndrome (PACS). Emerging evidence suggests that dysregulation of HIF-1α is a key driver of systemic inflammation, silent hypoxia, and pathological tissue remodeling in both the acute and post-acute phases of the disease. This scoping review was conducted following PRISMA-ScR guidelines and registered in INPLASY. It involved a literature search in Scopus and PubMed, supplemented by manual reference screening, with study selection facilitated by Rayyan software. Our analysis clarifies the dual role of HIF-1α, which may either worsen inflammatory responses and viral persistence or support adaptive mechanisms that reduce cellular damage. The potential for targeting HIF-1α therapeutically in COVID-19 is complex, requiring further investigation to clarify its precise role and translational applications. This review deepens the molecular understanding of SARS-CoV-2-induced cellular and tissue dysfunction in hypoxia, offering insights for improving clinical management strategies and addressing long-term sequelae. Full article
(This article belongs to the Special Issue Novel Insights into Molecular Mechanisms of Pulmonary Pathology)
Show Figures

Figure 1

22 pages, 2370 KiB  
Review
Curcumin Modulation of the Gut–Brain Axis for Neuroinflammation and Metabolic Disorders Prevention and Treatment
by Miriam Cerullo, Federica Armeli, Beatrice Mengoni, Martina Menin, Maria Luisa Crudeli and Rita Businaro
Nutrients 2025, 17(9), 1430; https://doi.org/10.3390/nu17091430 - 24 Apr 2025
Viewed by 2698
Abstract
Curcumin, a polyphenolic compound derived from Curcuma longa, has gained significant attention for its potential therapeutic benefits, particularly counteracting inflammation, oxidative stress, and metabolic disorders. Its chemical structure, featuring conjugated double bonds between two aromatic rings, allows it to act as an electron [...] Read more.
Curcumin, a polyphenolic compound derived from Curcuma longa, has gained significant attention for its potential therapeutic benefits, particularly counteracting inflammation, oxidative stress, and metabolic disorders. Its chemical structure, featuring conjugated double bonds between two aromatic rings, allows it to act as an electron donor, thereby mitigating free radical formation. Despite its poor solubility in water, curcumin is stable in acidic environments and undergoes significant metabolism in both the liver and the gut. Intestinal microbiota, particularly at the colon level, further metabolizes curcumin into several derivatives, including dihydrocurcumin and tetrahydrocurcumin, which exhibit antioxidant and anti-inflammatory properties. Studies suggest that curcumin can reduce body mass index (BMI) and improve other body composition parameters, especially when used in combination with lifestyle changes, though its bioavailability is low due to its rapid metabolism and the resulting low blood concentration. In obesity, dysfunctional adipose tissue remodeling and chronic inflammation play critical roles in the development of metabolic complications. Curcumin’s anti-inflammatory properties are related to the inhibition of the NF-κB pathway, leading to the reduction in inflammatory markers in adipocytes and macrophages. Additionally, curcumin modulates oxidative stress by activating the NRF2 pathway, enhancing cellular antioxidant defenses. Emerging evidence also supports curcumin’s potential in improving gut health by modulating microbiota composition, enhancing intestinal barrier function, and reducing systemic inflammation. This interaction with the gut–brain axis highlights the broader implications of curcumin in neuroprotection, as it positively affects cognitive function and mitigates neuroinflammation in neurodegenerative diseases like Alzheimer’s. disease. Thus, curcumin holds promise as a multifaceted agent in the management of obesity and associated diseases. Full article
Show Figures

Graphical abstract

25 pages, 2742 KiB  
Article
Combination of Exhaust Gas Fermentation Effluent and Dairy Wastewater for Microalgae Production: Effect on Growth and FAME Composition of Chlorella sorokiniana
by Elena Mazzocchi, Giulia Usai, Valeria Agostino, Silvia Fraterrigo Garofalo, Eugenio Pinton, Candido Fabrizio Pirri, Barbara Menin and Alessandro Cordara
Microorganisms 2025, 13(5), 961; https://doi.org/10.3390/microorganisms13050961 - 23 Apr 2025
Viewed by 699
Abstract
Microalgae cultivation in wastewater is a promising strategy for reducing nutrient loads and generating biomass that can be further exploited. Although microalgae grown under such conditions are not suitable for high-value applications, the resulting biomass can still be valuable for uses such as [...] Read more.
Microalgae cultivation in wastewater is a promising strategy for reducing nutrient loads and generating biomass that can be further exploited. Although microalgae grown under such conditions are not suitable for high-value applications, the resulting biomass can still be valuable for uses such as biofuels, biofertilizers, or animal feed. In this study, Chlorella sorokiniana was cultivated in dairy wastewater and, to the best of our knowledge, for the first time in a spent effluent from gas fermentation, to assess its potential as a sustainable growth medium. Growth kinetics and biomass productivity were evaluated at different dilution ratios, and it was found that high concentrations of ammonium and hexanol in undiluted effluents were inhibitory, while an optimized 50:50 dilution led to the highest biomass accumulation (1.96 g L−1) and productivity (0.5 g L−1 d−1) of C. sorokiniana. This strategy significantly reduced the nitrogen (100%), phosphate (100%), sulfate (68%), and carbon (61%) contents, demonstrating effective bioremediation activity. Furthermore, the fatty acid profile revealed an increased polyunsaturated fatty acid fraction, enhancing the potential of C. sorokiniana biomass as a feed supplement. Overall, contributing to the circular bioeconomy, this approach is scalable and cost-effective, reducing freshwater and chemical dependency in microalgae biomass production. Full article
Show Figures

Graphical abstract

13 pages, 550 KiB  
Review
Prognostic Impact of Phenotypic and Genetic Features of Pancreatic Malignancies
by Mikhail B. Potievskiy, Lidia A. Nekrasova, Ivan V. Korobov, Ekaterina A. Bykova, Ruslan I. Moshurov, Pavel V. Sokolov, Peter A. Shatalov, Natalia A. Falaleeva, Leonid O. Petrov, Vladimir S. Trifanov, Sergey A. Ivanov, Peter V. Shegai and Andrei D. Kaprin
Life 2025, 15(4), 635; https://doi.org/10.3390/life15040635 - 11 Apr 2025
Cited by 1 | Viewed by 835
Abstract
Pancreatic cancer is a tumor with a poor prognosis, and improving its survival outcomes remains a formidable challenge, requiring a multidisciplinary approach that integrates innovative surgical and pharmacological strategies, guided by molecular and genetic insights. The pathomorphological and genetic characteristics of pancreatic cancer, [...] Read more.
Pancreatic cancer is a tumor with a poor prognosis, and improving its survival outcomes remains a formidable challenge, requiring a multidisciplinary approach that integrates innovative surgical and pharmacological strategies, guided by molecular and genetic insights. The pathomorphological and genetic characteristics of pancreatic cancer, reflected in morphological, immunohistochemical, and serological marker expression, reveal key patterns of tumor genotypic changes during carcinogenesis, aiding in prognostic evaluation and clinical strategy development. The mutational profile of pancreatic tumors is quite heterogeneous and diverse in terms of mutated genes, including in relation to morphological subtypes, but certain patterns have been identified as a result of studies. Pancreatic adenocarcinoma, for instance, is frequently driven by mutations regulating cell division (KRAS). The disease prognosis often depends on the morphological subtype and tumor microenvironment. Neuroendocrine tumors of the pancreas are characterized by a number of pathogenetic features that distinguish them from adenocarcinomas. Thus, neuroendocrine tumors are characterized by mutations of the MENIN protein, which prevents cells from entering the mitosis phase by stimulating the expression of cell cycle regulators. Thus, epithelial and neuroendocrine malignancies of the pancreas differ in immunohistochemical and genetic features, but there are similar mechanisms of pathogenesis, such as BRCA1 and BRCA2 gene mutations, impaired expression of p53 antioncogene, and HIF-2α and mTOR receptor mutations. The predictive impact of serological markers, such as CA 19-9 and CEA, offers insights into tumor metastasis and long-term outcomes, emphasizing the need for personalized therapeutic strategies. Tailoring treatments based on individual molecular profiles holds promise for improving prognosis, as the genetic landscape of pancreatic tumors varies significantly between patients. This underscores the importance of a systematic, patient-specific approach that addresses tumor heterogeneity, resistance mechanisms, and the molecular underpinnings of carcinogenesis. Full article
(This article belongs to the Section Genetics and Genomics)
Show Figures

Figure 1

31 pages, 1305 KiB  
Review
Recent Developments in Differentiation Therapy of Acute Myeloid Leukemia
by Ugo Testa, Germana Castelli and Elvira Pelosi
Cancers 2025, 17(7), 1141; https://doi.org/10.3390/cancers17071141 - 28 Mar 2025
Cited by 1 | Viewed by 1121
Abstract
Acute myeloid leukemia (AML) is characterized by the clonal expansion of myeloid progenitors blocked at various stages of their differentiation process, and drugs that bypass this differentiation block are therapeutically efficient, as shown by retinoic acid and arsenic trioxide in acute promyelocytic leukemia. [...] Read more.
Acute myeloid leukemia (AML) is characterized by the clonal expansion of myeloid progenitors blocked at various stages of their differentiation process, and drugs that bypass this differentiation block are therapeutically efficient, as shown by retinoic acid and arsenic trioxide in acute promyelocytic leukemia. However, the successful application of differentiation therapy in APL has not translated into clinical benefit for other non-APL subtypes of AML, in which intensive chemotherapy regimens represent the standard of care. However, the development of molecular studies has led to the identification of therapeutic targets (such as mutated proteins and deregulated pathways) and has led to the generation of a new category of specific pharmacologic agents. Some of these agents, such as inhibitors of mutant isocitrate dehydrogenase (IDH1 and IDH2), lysine-specific demethylase-1 (LSD1), and Menin, have shown the capacity to induce leukemic cell differentiation and with significant therapeutic efficacy. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Figure 1

19 pages, 980 KiB  
Review
Menin Inhibitors: New Targeted Therapies for Specific Genetic Subtypes of Difficult-to-Treat Acute Leukemias
by Pasquale Niscola, Valentina Gianfelici, Marco Giovannini, Daniela Piccioni, Carla Mazzone and Paolo de Fabritiis
Cancers 2025, 17(1), 142; https://doi.org/10.3390/cancers17010142 - 4 Jan 2025
Cited by 1 | Viewed by 3233
Abstract
Menin (MEN1) is a well-recognized powerful tumor promoter in acute leukemias (AL) with KMT2A rearrangements (KMT2Ar, also known as MLL) and mutant nucleophosmin 1 (NPM1m) acute myeloid leukemia (AML). MEN1 is essential for sustaining leukemic transformation due to its interaction with wild-type KMT2A [...] Read more.
Menin (MEN1) is a well-recognized powerful tumor promoter in acute leukemias (AL) with KMT2A rearrangements (KMT2Ar, also known as MLL) and mutant nucleophosmin 1 (NPM1m) acute myeloid leukemia (AML). MEN1 is essential for sustaining leukemic transformation due to its interaction with wild-type KMT2A and KMT2A fusion proteins, leading to the dysregulation of KMT2A target genes. MEN1 inhibitors (MIs), such as revumenib, ziftomenib, and other active small molecules, represent a promising new class of therapies currently under clinical development. By disrupting the MEN1-KMT2Ar complex, a group of proteins involved in chromatin remodeling, MIs induce apoptosis and differentiation AL expressing KMT2Ar or NPM1m AML. Phase I and II clinical trials have evaluated MIs as standalone treatments and combined them with other synergistic drugs, yielding promising results. These trials have demonstrated notable response rates with manageable toxicities. Among MIs, ziftomenib received orphan drug and breakthrough therapy designations from the European Medicines Agency in January 2024 and the Food and Drug Administration (FDA) in April 2024, respectively, for treating R/R patients with NPM1m AML. Additionally, in November 2024, the FDA approved revumenib for treating R/R patients with KMT2Ar-AL. This review focuses on the pathophysiology of MI-sensitive AL, primarily AML. It illustrates data from clinical trials and discusses the emergence of resistance mechanisms. In addition, we outline future directions for the use of MIs and emphasize the need for further research to fully realize the potential of these novel compounds, especially in the context of specific genetic subtypes of challenging AL. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Figure 1

15 pages, 2071 KiB  
Article
Unveiling the Complexity of KMT2A Rearrangements in Acute Myeloid Leukemias with Optical Genome Mapping
by Sandrine A. Lacoste, Vanessa Gagnon, François Béliveau, Sylvie Lavallée, Vanessa Collin and Josée Hébert
Cancers 2024, 16(24), 4171; https://doi.org/10.3390/cancers16244171 - 14 Dec 2024
Cited by 2 | Viewed by 1668
Abstract
Background: KMT2A rearrangements are major genetic entities in the classification of acute myeloid leukemias (AMLs), but their diverse and frequently cryptic nature makes their detection and characterization challenging. Karyotypic anomalies at the KMT2A locus and/or abnormal KMT2A Fluorescence in situ hybridization (FISH) [...] Read more.
Background: KMT2A rearrangements are major genetic entities in the classification of acute myeloid leukemias (AMLs), but their diverse and frequently cryptic nature makes their detection and characterization challenging. Karyotypic anomalies at the KMT2A locus and/or abnormal KMT2A Fluorescence in situ hybridization (FISH) results strongly indicate a KMT2A fusion, but the identification of the translocation partner gene often requires further investigation. KMT2A partial tandem duplications (PTDs), on the other hand, are undetectable by standard cytogenetics methods. Methods: We herein report the optical genome mapping (OGM) analysis of 38 AML samples: 12 cryptic/hard-to-characterize KMT2A fusions, 20 KMT2A-PTDs and 6 cases with no KMT2A anomaly. Results: In all the fusion cases, the rearrangement between 5’KMT2A and the 3’partner gene was identified as a translocation t(v;11q23.3)(v;118479068), and the analysis of co-occurring variants elucidated the formation of the rearrangement. The KMT2A variants detected in the KMT2A-PTD cases were surprisingly diverse. Combined with RNAseq data, OGM analysis identified 9 distinct in-frame KMT2A-PTD variants among the 20 cases analyzed. Conclusions: With the clinical development of menin inhibitors for the treatment of patients with KMT2A-rearranged acute leukemias, the characterization of these rearrangements is of utmost importance. Our results suggest that OGM is a promising tool for accurate genetic diagnosis in this context. Full article
(This article belongs to the Section Molecular Cancer Biology)
Show Figures

Figure 1

15 pages, 790 KiB  
Review
Synergistic Strategies for KMT2A-Rearranged Leukemias: Beyond Menin Inhibitor
by Sandra Cantilena, Mohamed AlAmeri, Noelia Che, Owen Williams and Jasper de Boer
Cancers 2024, 16(23), 4017; https://doi.org/10.3390/cancers16234017 - 29 Nov 2024
Cited by 1 | Viewed by 2629
Abstract
KMT2A-rearranged leukemias are a highly aggressive subset of acute leukemia, characterized by poor prognosis and frequent relapses despite intensive treatment. Menin inhibitors, which target the critical KMT2A–menin interaction driving leukemogenesis, have shown promise in early clinical trials. However, resistance to these inhibitors, often [...] Read more.
KMT2A-rearranged leukemias are a highly aggressive subset of acute leukemia, characterized by poor prognosis and frequent relapses despite intensive treatment. Menin inhibitors, which target the critical KMT2A–menin interaction driving leukemogenesis, have shown promise in early clinical trials. However, resistance to these inhibitors, often driven by menin mutations or alternative oncogenic pathways, remains a significant challenge. This review explores combination therapies aimed at overcoming resistance and improving patient outcomes. Potential strategies include inhibiting DOT1L, a histone methyltransferase essential for KMT2A-driven transcription, and BRD4, a regulator of transcriptional super-enhancers. Additionally, targeting MYC, a key oncogene frequently upregulated in KMT2A-rearranged leukemia, offers another approach. Direct inhibition of KMT2A-fusion proteins and c-MYB, a transcription factor critical for leukemic stem cell maintenance, is also explored. By integrating these diverse strategies, we propose a comprehensive therapeutic paradigm that targets multiple points of the leukemic transcriptional and epigenetic network. These combination approaches aim to disrupt key oncogenic pathways, reduce resistance, and enhance treatment efficacy, ultimately providing more durable remissions and improved survival for patients with KMT2A-rearranged leukemias. Full article
(This article belongs to the Special Issue The Clinical Trials and Management of Acute Myeloid Leukemia)
Show Figures

Graphical abstract

15 pages, 2643 KiB  
Article
Patient-Specific Circulating Tumor DNA for Monitoring Response to Menin Inhibitor Treatment in Preclinical Models of Infant Leukemia
by Louise Doculara, Kathryn Evans, J. Justin Gooding, Narges Bayat and Richard B. Lock
Cancers 2024, 16(23), 3990; https://doi.org/10.3390/cancers16233990 - 28 Nov 2024
Viewed by 1090
Abstract
Background: In infant KMT2A (MLL1)-rearranged (MLL-r) acute lymphoblastic leukemia (ALL), early relapse and treatment response are currently monitored through invasive repeated bone marrow (BM) biopsies. Circulating tumor DNA (ctDNA) in peripheral blood (PB) provides a minimally invasive alternative, allowing for more [...] Read more.
Background: In infant KMT2A (MLL1)-rearranged (MLL-r) acute lymphoblastic leukemia (ALL), early relapse and treatment response are currently monitored through invasive repeated bone marrow (BM) biopsies. Circulating tumor DNA (ctDNA) in peripheral blood (PB) provides a minimally invasive alternative, allowing for more frequent disease monitoring. However, a poor understanding of ctDNA dynamics has hampered its clinical translation. We explored the predictive value of ctDNA for detecting minimal/measurable residual disease (MRD) and drug response in a patient-derived xenograft (PDX) model of infant MLL-r ALL. Methods: Immune-deficient mice engrafted with three MLL-r ALL PDXs were monitored for ctDNA levels before and after treatment with the menin inhibitor SNDX-50469. Results: The amount of ctDNA detected strongly correlated with leukemia burden during initial engraftment prior to drug treatment. However, following SNDX-50469 treatment, the leukemic burden assessed by either PB leukemia cells through flow cytometry or ctDNA levels through droplet digital polymerase chain reaction (ddPCR) was discrepant. This divergence could be attributed to the persistence of leukemia cells in the spleen and BM, highlighting the ability of ctDNA to reflect disease dynamics in key leukemia infiltration sites. Conclusions: Notably, ctDNA analysis proved to be a superior predictor of MRD compared to PB assessment alone, especially in instances of low disease burden. These findings highlight the potential of ctDNA as a sensitive biomarker for monitoring treatment response and detecting MRD in infant MLL-r ALL. Full article
(This article belongs to the Section Pediatric Oncology)
Show Figures

Figure 1

19 pages, 2205 KiB  
Article
An Ultra-Fast Validated Green UPLC-MS/MS Approach for Assessing Revumenib in Human Liver Microsomes: In Vitro Absorption, Distribution, Metabolism, and Excretion and Metabolic Stability Evaluation
by Mohamed W. Attwa, Ali S. Abdelhameed and Adnan A. Kadi
Medicina 2024, 60(12), 1914; https://doi.org/10.3390/medicina60121914 - 21 Nov 2024
Cited by 7 | Viewed by 1420
Abstract
Background and Objectives: Revumenib (SNDX-5613) is a powerful and specific inhibitor of the menin–KMT2A binding interaction. It is a small molecule that is currently being researched to treat KMT2A-rearranged (KMT2Ar) acute leukemias. Revumenib (RVB) has received Orphan Drug Designation from the US FDA [...] Read more.
Background and Objectives: Revumenib (SNDX-5613) is a powerful and specific inhibitor of the menin–KMT2A binding interaction. It is a small molecule that is currently being researched to treat KMT2A-rearranged (KMT2Ar) acute leukemias. Revumenib (RVB) has received Orphan Drug Designation from the US FDA for treating patients with AML. It has also been granted Fast Track designation by the FDA for treating pediatric and adult patients with R/R acute leukemias that have a KMT2Ar or NPM1 mutation. Materials and Methods: The target of this research was to create a fast, precise, green, and extremely sensitive UPLC-MS/MS technique for the estimation of the RVB level in human liver microsomes (HLMs), employing an ESI source. The validation procedures were carried out in accordance with the bioanalytical technique validation requirements established by the US Food and Drug Administration that involve linearity, selectivity, precision, accuracy, stability, matrix effect, and extraction recovery. The outcome data of the validation features of the UPLC-MS/MS approach were acceptable according to FDA guidelines. RVB parent ions were formed in the positive ESI source and its two fragment ions were estimated employing multiple reaction monitoring (MRM) mode. The separation of RVB and encorafenib was achieved using a C8 column (2.1 mm, 50 mm, and 3.5 µm) and isocratic mobile phase. Results: The RVB calibration curve linearity ranged from 1 to 3000 ng/mL (y = 0.6515x − 0.5459 and R2 = 0.9945). The inter-day precision and accuracy spanned from −0.23% to 11.33%, while the intra-day precision and accuracy spanned from −0.88% to 11.67%, verifying the reproducibility of the UPLC-MS/MS analytical technique. The sensitivity of the developed methodology demonstrated its capability to quantify RVB levels at an LOQ of 0.96 ng/mL. The AGREE score was 0.77, confirming the greenness of the current method. The low in vitro t1/2 (14.93 min) and high intrinsic clearance (54.31 mL/min/kg) of RVB revealed that RVB shares similarities with medications that have a high extraction ratio. Conclusions: The present LC-MS/MS approach is considered the first analytical approach with the application of metabolic stability assessment for RVB estimation in HLMs. These methods are essential for advancing the development of new pharmaceuticals, particularly in enhancing metabolic stability. Full article
(This article belongs to the Special Issue Acute Myeloid Leukemia: Update on Diagnosis, Therapy, and Monitoring)
Show Figures

Figure 1

Back to TopTop