ChemMedChemPub Date : 2025-09-17DOI: 10.1002/cmdc.202500209
Chelsi M Almodóvar-Rivera, Ira Tandon, Ramesh Mudududdla, Paulina N Esguerra, Kevin Lucio-Acero, Weiping Tang
{"title":"Development of a Partial Proteolysis Targeting Chimera Library Based on Achiral Cereblon E3 Ligase Ligands and its Application for Bruton's Tyrosine Kinase Degraders.","authors":"Chelsi M Almodóvar-Rivera, Ira Tandon, Ramesh Mudududdla, Paulina N Esguerra, Kevin Lucio-Acero, Weiping Tang","doi":"10.1002/cmdc.202500209","DOIUrl":"10.1002/cmdc.202500209","url":null,"abstract":"<p><p>Proteolysis targeting chimeras (PROTACs) offer a promising therapeutic approach by leveraging the ubiquitin-proteasome system (UPS) to degrade target proteins. These heterobifunctional molecules consist of a target protein ligand, an E3 ligase ligand, and a linker. Among the limited E3 ligase ligands available, cereblon (CRBN) ligands are the most widely used. However, the stability and racemization of current chiral CRBN ligands pose challenges for developing CRBN-recruiting PROTAC therapeutics. Herein, a partial PROTAC library is reported incorporating an aldehyde motif and various linkers into previously developed achiral phenyl dihydrouracil CRBN ligands, which offer improved stability and eliminate racemization issues. This library enables the rapid generation of fully functional PROTACs targeting Bruton's tyrosine kinase (BTK) by coupling the aldehyde motif with a hydrazide-containing BTK ligand. Initial HiBiT assay (Promega) screening identifies nine hits capable of significant BTK degradation, with compound B1 emerging as the most potent degrader. A stable amide bioisostere, AM-B1, is further developed, which induces significant antiproliferation and BTK degradation. Mechanistic studies confirm BTK degradation via the UPS. This study highlights the development of an achiral CRBN-based partial PROTAC library and demonstrates a two-stage strategy for rapid PROTAC development against BTK.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202500209"},"PeriodicalIF":3.4,"publicationDate":"2025-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12471865/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145079185","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Rational Design and Antimycobacterial Evaluation of Aryl Sulfonamide-Linked Isoniazid Hydrazones Against Mycobacterium Tuberculosis.","authors":"Mukanda Gedeon Kadima, Sahil Mishra, Gobind Kumar, Pule Seboletswe, Afsana Kajee, Ankit, Françoise Roquet-Banères, Maëlle Foubert, Laurent Kremer, Rajshekhar Karpoormath, Parvesh Singh","doi":"10.1002/cmdc.202500398","DOIUrl":"https://doi.org/10.1002/cmdc.202500398","url":null,"abstract":"<p><p>Despite significant advancements in antituberculosis (TB) drug discovery, considerable scope remains for novel therapeutic development. Molecular hybridization represents a promising strategy for generating new anti-TB agents. In this study, in silico molecular docking is employed to design novel isoniazid-sulfonamide hybrids connected via a hydrazone bridge, designated as series 7j-r and 8a-i. Docking analysis reveals that these compounds interact significantly with the active site of InhA, particularly engaging the catalytic triad residues Y158, F149, and K165, as well as the cofactor NAD. Subsequently, both series are synthesized and evaluated against Mycobacterium tuberculosis. Generally, compounds from both series (7 and 8) exhibit enhanced activity compared to their precursors. Notably, compound 8a demonstrated approximately twofold greater potency ( minimum inhibitory concentration (MIC) = 0.156 µg mL<sup>-1</sup>) with respect to compound 7j (MIC = 0.313 µg mL<sup>-1</sup>). However, these compounds lose efficacy against INH-resistant M. tuberculosis strains harboring katG mutations and remain ineffective against multidrug-resistant and extensively drug-resistant strains of M. tuberculosis. Encouragingly, the tested compounds exhibit little cytotoxicity against the THP-1 human monocytic cell line at a concentration of 20 µg mL<sup>-1</sup>. Additionally, the structural stability studies using <sup>1</sup>H NMR confirm the structural integrity of these compounds. Overall, these molecular hybrids are promising for further development as anti-TB agents after relevant structural optimizations.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202500398"},"PeriodicalIF":3.4,"publicationDate":"2025-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145079183","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ChemMedChemPub Date : 2025-09-14DOI: 10.1002/cmdc.202500600
Francisca Lopes, Gábor Girst, Rafael Rincón, Ricardo J F Ferreira, Lídia M Gonçalves, Lucília Saraiva, Hui-Chun Wang, Muriel Cuendet, Attila Hunyadi, Maria M M Santos
{"title":"Hybrid Molecules of p53 Activators and Protoflavones to Target Multiple Myeloma.","authors":"Francisca Lopes, Gábor Girst, Rafael Rincón, Ricardo J F Ferreira, Lídia M Gonçalves, Lucília Saraiva, Hui-Chun Wang, Muriel Cuendet, Attila Hunyadi, Maria M M Santos","doi":"10.1002/cmdc.202500600","DOIUrl":"https://doi.org/10.1002/cmdc.202500600","url":null,"abstract":"<p><p>Multiple myeloma is a rare blood cancer that develops from abnormal plasma cells in the bone marrow. Treatment of multiple myeloma remains an enormous challenge. In this work, hybrid compounds are developed and studied for their potential use against multiple myeloma. The compounds are designed to act by a dual mechanism of action, activation of the p53 pathway, and inhibition of the ataxia telangiectasia and Rad3-related protein (ATR). To evaluate the selectivity for the p53 pathway, the compounds are first evaluated in an isogenic pair of HCT116 colon cancer cell lines, with and without p53, and in two breast cancer cell lines expressing different forms of p53. Then, the growth inhibitory effect of the hybrid compounds is tested against the multiple myeloma cell lines RPMI 8226 (mutant p53) and MM.1S (wild-type p53). At the same time, compound 15 is confirmed to inhibit doxorubicin- but not UV-induced DNA damage response via the ATR/Chk1 signaling pathway. The hybrids show lower IC<sub>50</sub> values compared to the fragments alone, highlighting the potential for using hybrid molecules containing two pharmacophores with complementary activities. These results suggest that novel hybrid molecules may serve as new leads against multiple myeloma.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202500600"},"PeriodicalIF":3.4,"publicationDate":"2025-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145058129","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Targeting Plasmodium falciparum Single-Stranded DNA-Binding Protein: Discovery of New Scaffold Compounds Effective against Drug-Sensitive and Artemisinin-Resistant Strains.","authors":"Biswajit Naik, Preshita Bhalerao, Shashank Shekhar, Stephy A Varghese, Sweta Makwana, Chandi C Mandal, Tarun Kumar Bhatt, Suman Kumar Dhar, Dhaneswar Prusty","doi":"10.1002/cmdc.202500282","DOIUrl":"https://doi.org/10.1002/cmdc.202500282","url":null,"abstract":"<p><p>Resistance to frontline antimalarials by malaria parasites is a major concern, prompting the search for novel antimalarial compounds targeting new drug targets. One promising target is the single-stranded DNA-binding protein (PfSSB) of Plasmodium falciparum, crucial for DNA replication process in apicoplast, an essential organelle. Using an integrative approach, PPG and 9-HPF are identified, which inhibit the DNA binding property of PfSSB and exhibit antimalarial activity. Computational analyses reveal that both compounds possess strong binding affinity and favorable drug-like properties. Biolayer interferometry assay and the gel retardation assays demonstrate that they have a stronger binding affinity and disrupt PfSSB's ssDNA binding ability. PPG and 9-HPF show antimalarial activity against drug-sensitive P. falciparum (3D7) with IC<sub>50</sub> values of 54.95 and 9.13 μM, respectively, and they can also effectively inhibit artemisinin-resistant P. falciparum (C580Y) with a resistance index of 0.79 and 1.26, respectively. MTT cytotoxicity assays confirm that both compounds are nontoxic, and structural similarity analysis indicates that they are distinct from existing antimalarials, reducing cross-resistance risk. Additionally, virtual screening of 59,807 compounds with similar structures to PPG and 9-HPF identifies candidates with higher affinities for PfSSB. This study identifies PPG and 9-HPF as structurally distinct antimalarials with the potential to combat drug-resistant P. falciparum.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202500282"},"PeriodicalIF":3.4,"publicationDate":"2025-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145058146","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ChemMedChemPub Date : 2025-09-14DOI: 10.1002/cmdc.202500350
Ledy De-la-Cruz-Martínez, Rosendo Martínez-Arellano, Mitzi López-Sánchez, José G Alvarado-Rodríguez, Jesús Martin Torres-Valencia, David Equihua-González, Julio-César Almanza-Pérez, Jaime Pérez-Villanueva, Martín González-Andrade, José C Páez-Franco, Francisco Cortés-Benítez
{"title":"Impact of C18 Epimerization of Indole- and Pyrazole-Fused 18β-Glycyrrhetinic Acid Derivatives on PTP1B and TCPTP Inhibitory Activity: Synthesis, In Vitro, and In Silico Studies.","authors":"Ledy De-la-Cruz-Martínez, Rosendo Martínez-Arellano, Mitzi López-Sánchez, José G Alvarado-Rodríguez, Jesús Martin Torres-Valencia, David Equihua-González, Julio-César Almanza-Pérez, Jaime Pérez-Villanueva, Martín González-Andrade, José C Páez-Franco, Francisco Cortés-Benítez","doi":"10.1002/cmdc.202500350","DOIUrl":"https://doi.org/10.1002/cmdc.202500350","url":null,"abstract":"<p><p>Protein tyrosine phosphatase 1B (PTP1B) is crucial for negatively regulating the canonical insulin and leptin signaling pathways. This enzyme is a validated target for treating various disorders, including diabetes and obesity. However, to date, no PTP1B inhibitors have been approved for use. In earlier studies, we developed two modified versions of 18β-glycyrrhetinic acid (18β-GA) called FC-114 and FC-122, which showed better inhibitory PTP1B activity than ursolic acid, a well-known inhibitor. To develop even stronger inhibitors, we looked at another compound, 18α-glycyrrhetinic acid (18α-GA), which is more potent than 18β-GA. Thus, in this study, we aimed to synthesize the analogs 18epi-FC114 (3c) and 18epi-FC-122 (5c). These compounds were prepared with and without the carbonyl group at C11. The results showed that converting 18β-H to 18α-H, as well as the absence of the 11-carbonyl group, negatively impacted the PTP1B inhibitory activity. However, the synthesized compounds exhibited an uncompetitive type of inhibition toward PTP1B and did not inhibit the TCPTP enzyme. Molecular docking and dynamics simulations suggest that the inversion of 18β-H pushes the 30-COOH group away, disrupting interactions at the C-terminal site of PTP1B<sub>1-400</sub>. Additionally, the absence of the 11-carbonyl group positions the compounds unfavorably, limiting critical interactions in the same region.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202500350"},"PeriodicalIF":3.4,"publicationDate":"2025-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145058169","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ChemMedChemPub Date : 2025-09-14DOI: 10.1002/cmdc.202500333
Rosa Giugliano, Carla Zannella, Annalisa Chianese, Clementina Acconcia, Alessandra Monti, Roberta Della Marca, Ugo Pagnini, Serena Montagnaro, Nunzianna Doti, Carla Isernia, Massimiliano Galdiero, Filomena Fiorito, Luigi Russo, Valentina Iovane, Anna De Filippis
{"title":"Pantinin-Derived Peptides against Veterinary Herpesviruses: Activity and Structural Characterization.","authors":"Rosa Giugliano, Carla Zannella, Annalisa Chianese, Clementina Acconcia, Alessandra Monti, Roberta Della Marca, Ugo Pagnini, Serena Montagnaro, Nunzianna Doti, Carla Isernia, Massimiliano Galdiero, Filomena Fiorito, Luigi Russo, Valentina Iovane, Anna De Filippis","doi":"10.1002/cmdc.202500333","DOIUrl":"https://doi.org/10.1002/cmdc.202500333","url":null,"abstract":"<p><p>Animal viral infections represent a growing public health concern, as animals serve as reservoirs for pathogens, threatening food safety, biodiversity, and human health. In response, novel antiviral strategies are urgently needed. This study investigates the antiviral activity and structural properties of two antimicrobial peptides, pantinin-1 and pantinin-2, both derived from the venom of the scorpion Pandinus imperator, against caprine herpesvirus 1 (CpHV-1) and bovine herpesvirus 1 (BoHV-1). The results obtained from the plaque reduction assay and the quantitative real-time polymerase chain reaction (PCR) indicate that synthetic pantinin-mimetic peptides exhibited potent antiviral effects at concentrations ranging from 6-25 µM, impairing viral infectivity through direct virucidal action and inhibition of the viral entry and fusion with host cell. To characterize their structural behavior, nuclear magnetic resonance spectroscopy is performed in aqueous and membrane-mimetic environments (trifluoroethanol (TFE)/H<sub>2</sub>O). In aqueous solution, both peptides predominantly adopted random coil conformations, with pantinin-2 showing greater secondary structure propensity. In TFE/H<sub>2</sub>O, both peptides transitioned to α-helical structures, which are often associated with membrane interaction and antiviral activity. These findings demonstrate that pantinin-1 and pantinin-2 possess promising antiviral properties, supporting their potential development as therapeutic agents against herpesviruses and other animal viral infections.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202500333"},"PeriodicalIF":3.4,"publicationDate":"2025-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145058159","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ChemMedChemPub Date : 2025-09-14DOI: 10.1002/cmdc.202500577
Nam Q H Doan, Huyen T T Nguyen, Hoang-Thuc Huynh, Tuyen Ngoc Truong
{"title":"Curcumin Analogs Containing Pyrazole-Pyridine Hybrids as Novel Anticancer Agents: Synthesis, Cytotoxicity, Apoptosis Induction, and Molecular Modeling.","authors":"Nam Q H Doan, Huyen T T Nguyen, Hoang-Thuc Huynh, Tuyen Ngoc Truong","doi":"10.1002/cmdc.202500577","DOIUrl":"https://doi.org/10.1002/cmdc.202500577","url":null,"abstract":"<p><p>In recent years, curcumin analogs have not only demonstrated potent anticancer activities but have also addressed several limitations of curcumin itself, thereby remaining a promising focus of research within the scientific community. Building upon the findings of our previous studies, structural modifications of potent curcumin analogs fused with 1H-pyrazole are performed by applying the bioisosteric replacement strategy of a benzene ring with a pyridine ring, to develop a series of novel curcumin analogs containing pyrazole-pyridine hybrids (3a-4h) as promising anticancer agents. Among them, curcumin analog 4c emerges as the most potent compound, exhibiting the strongest cytotoxicity against various human cancer cell lines, including HepG2 (liver), MDA-MB-231 (breast), and A549 (lung), as well as significant apoptosis-inducing effects in HepG2 cells. Furthermore, compound 4c is predicted to possess a favorable physicochemical-pharmacokinetic-toxicological profile, as well as an effective binding mode at the colchicine-binding site of the α,β-tubulin heterodimer. Importantly, the bioisosteric replacement in compound 4c is found to exert beneficial effects on its anticancer activities, physicochemical-pharmacokinetic-toxicological properties, and binding affinity, in comparison with its parent compounds.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202500577"},"PeriodicalIF":3.4,"publicationDate":"2025-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145058174","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ChemMedChemPub Date : 2025-09-14DOI: 10.1002/cmdc.202500174
Sayed M Safwan, Devashish Mehta, Varsha Saini, Dolly Jain, Nikhil K Chourasiya, Amit Arora, Steffi Khatol, Mohit Singh, Vikas Verma, Avinash Bajaj
{"title":"Aspirin-Conjugated Cholic Acid Derivative Combats Polymicrobial Infections and Inflammation.","authors":"Sayed M Safwan, Devashish Mehta, Varsha Saini, Dolly Jain, Nikhil K Chourasiya, Amit Arora, Steffi Khatol, Mohit Singh, Vikas Verma, Avinash Bajaj","doi":"10.1002/cmdc.202500174","DOIUrl":"https://doi.org/10.1002/cmdc.202500174","url":null,"abstract":"<p><p>Treating wound infections caused by Gram-positive and Gram-negative bacteria, along with associated inflammation, remains challenging due to the limited therapeutic efficacy of conventional antibiotics. Leveraging the unique properties of cholic acid derivatives, such as their cationic nature, hydrophobicity, and structural adaptability, a novel aspirin-derived facile amphiphile based on cholic acid (amphiphile 1) is designed and synthesized. The investigations reveal that the amphiphile 1 functions as a bacterial membrane disruptor through interacting with key components such as lipoteichoic acid and lipopolysaccharide of bacterial membranes. It is observed that amphiphile 1 can degrade both monomicrobial and polymicrobial preformed biofilms originating from Gram-positive and Gram-negative bacteria. Animal studies demonstrate that hydrogel-based delivery of amphiphile 1 effectively mitigates bacterial infections and resolves bacterial-associated inflammation. Collectively, these findings underscore that amphiphile 1 is a putative therapeutic agent for addressing the challenges of polymicrobial infections.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202500174"},"PeriodicalIF":3.4,"publicationDate":"2025-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145058131","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ChemMedChemPub Date : 2025-09-14DOI: 10.1002/cmdc.202500470
Gaurav Sheth, Shailesh R Shah, Prabal Sengupta, Tushar Jarag, Sabbirhusen Chimanwala, Kalapatapu V V M Sairam, Vaibhav Jain, Rashmi Talwar, Avinash Dhanave, Mehul Raviya, Harendra Jha, Rajasekhar Reddy Chilakala, Trinadha Rao Chitturi
{"title":"Strategic Design and Development of Indole-Based Compounds as Potent malic Enzyme 3 Inhibitors for Pancreatic Tumor Therapy.","authors":"Gaurav Sheth, Shailesh R Shah, Prabal Sengupta, Tushar Jarag, Sabbirhusen Chimanwala, Kalapatapu V V M Sairam, Vaibhav Jain, Rashmi Talwar, Avinash Dhanave, Mehul Raviya, Harendra Jha, Rajasekhar Reddy Chilakala, Trinadha Rao Chitturi","doi":"10.1002/cmdc.202500470","DOIUrl":"https://doi.org/10.1002/cmdc.202500470","url":null,"abstract":"<p><p>Malic enzyme 3 (ME3) plays a critical role in the survival of SMAD4<sup>-/-</sup>/ME2<sup>-/-</sup> pancreatic ductal adenocarcinoma (PDAC) cells by supporting energy production and maintaining redox homeostasis. Therefore, targeting ME3 with small-molecule inhibitors presents a promising therapeutic strategy for PDAC patients with SMAD4/ME2 deletions. Building upon our previously developed ME3 inhibitor, a systematic exploration of the structure-activity relationship (SAR) was undertaken to identify novel chemotypes. This effort led to the discovery of a new series of indole-substituted piperazine carboxamides with potent ME3 inhibitory activity, among which compound 13 emerged to be the most effective in PDAC cell lines. Furthermore, the synergistic effects of the newly identified compound 13 with the mitogen-activated protein kinase kinase (MEK) inhibitor trametinib were evaluated on Hs766T cells which revealed a significant synergism of this combination.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202500470"},"PeriodicalIF":3.4,"publicationDate":"2025-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145058115","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Improving the Catalyst Efficiency for Hyperpolarization of Pyruvate Derivatives by Means of Hydrogenative PHIP.","authors":"Ginevra Di Matteo, Oksana Bondar, Carla Carrera, Eleonora Cavallari, Sumit Mishra, Francesca Reineri","doi":"10.1002/cmdc.202500379","DOIUrl":"https://doi.org/10.1002/cmdc.202500379","url":null,"abstract":"<p><p>Hyperpolarized pyruvate is the most widely used probe for metabolic imaging in magnetic resonance (MR). Parahydrogen induced polarization- side arm hydrogenation allows to generate it through the catalytic hydrogenation of pyruvate esters. Due to the transient nature of MR hyperpolarization and to the fact that in vivo applications require a high amount of hyperpolarized substrate, a concentrated product solution must be obtained in a few seconds, therefore a high catalyst concentration is needed. The homogeneous rhodium catalyst used for the reaction can be deactivated by the hydrogenation products (or substrates) and the substrate-to-catalyst ratio becomes even lower, especially for pyruvate esters. The addition of tris-phenyl phosphine to the hydrogenation mixture prevents the catalyst deactivation, when it is due to the hydrogenation product allyl pyruvate and the amount of catalyst needed to obtain a concentrated batch of hyperpolarized pyruvate ester has been reduced significantly. Following to hydrolysis and extraction of sodium pyruvate in aqueous phase, the concentration of the hyperpolarized metabolite has been increased to about 60 mM and <sup>13</sup>C-MRI experiments have been carried out using different dilution of the hyperpolarized metabolite in water.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202500379"},"PeriodicalIF":3.4,"publicationDate":"2025-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145038830","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}