Veronika A Szalai, Christina Bergonzo, Rachel B Lyon, Zvi Kelman, Thomas Schmidt, Alexander Grishaev
Antibody-based pharmaceuticals are the leading biologic drug platform (> $75B/year).[1] Despite a wealth of information collected on them, there is still a lack of knowledge on their inter-domain structural distributions, which impedes innovation and development. To address this measurement gap, we have developed a new methodology to derive biomolecular structure ensembles from distance distribution measurements via a library of tagged proteins bound to an unlabeled and otherwise unmodified target biologic. We have employed the NIST monoclonal antibody (NISTmAb) reference material as our development platform for use with spin-labeled affinity protein (SLAP) reagents. Using double electron-electron resonance (DEER) spectroscopy, we have determined inter-spin distance distributions in SLAP complexes of both the isolated Fc domain and the intact NISTmAb. Our SLAP reagents offer a general and extendable technology, compatible with any non-isotopically labeled immunoglobulin G class mAb. Integrating molecular simulations with the DEER and solution X-ray scattering measurements, we enable simultaneous determination of structural distributions and dynamics of mAb-based biologics.
{"title":"Structure and Dynamics of Monoclonal Antibody Domains Using Spins, Scattering, and Simulations.","authors":"Veronika A Szalai, Christina Bergonzo, Rachel B Lyon, Zvi Kelman, Thomas Schmidt, Alexander Grishaev","doi":"10.1002/cmdc.202400917","DOIUrl":"10.1002/cmdc.202400917","url":null,"abstract":"<p><p>Antibody-based pharmaceuticals are the leading biologic drug platform (> $75B/year).<sup>[1]</sup> Despite a wealth of information collected on them, there is still a lack of knowledge on their inter-domain structural distributions, which impedes innovation and development. To address this measurement gap, we have developed a new methodology to derive biomolecular structure ensembles from distance distribution measurements via a library of tagged proteins bound to an unlabeled and otherwise unmodified target biologic. We have employed the NIST monoclonal antibody (NISTmAb) reference material as our development platform for use with spin-labeled affinity protein (SLAP) reagents. Using double electron-electron resonance (DEER) spectroscopy, we have determined inter-spin distance distributions in SLAP complexes of both the isolated Fc domain and the intact NISTmAb. Our SLAP reagents offer a general and extendable technology, compatible with any non-isotopically labeled immunoglobulin G class mAb. Integrating molecular simulations with the DEER and solution X-ray scattering measurements, we enable simultaneous determination of structural distributions and dynamics of mAb-based biologics.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202400917"},"PeriodicalIF":3.6,"publicationDate":"2025-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142968991","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}
Helge Vatheuer, Oscar Palomino-Hernández, Janis Müller, Phillip Galonska, Serghei Glinca, Paul Czodrowski
Protonation states serve as an essential molecular recognition motif for biological processes. Their correct consideration is key to successful drug design campaigns, since chemoinformatic tools usually deal with default protonation states of ligands and proteins and miss atypical protonation states. The protonation pattern for the Endothiapepsin/PepstatinA (EP/pepA) complex is investigated using different dry lab and wet lab techniques. ITC experiments revealed an uptake of more than one mole of protons upon pepA binding to EP. Since these experiments were performed at physiological conditions (and not at pH=4.6 at which a large variety of crystal structures is available), a novel crystal structure at pH=7.6 was determined. This crystal structure showed that only modest structural changes occur upon increasing the pH value. This lead to computational studies Poisson-Boltzmann calculations and constant pH MD simulation to reveal the exact location of the protonation event. Both computational studies could reveal a significant pKa shift resulting in non-default protonation state and that the catalytic dyad is responsible for the uptake of protons. This study shows that assessing protonation states for two separate systems (protein and ligand) might result in the incorrect assignment of protonation states and hence incorrect calculation of binding energy.
{"title":"Protonation Effects in Protein-Ligand Complexes - A Case Study of Endothiapepsin and Pepstatin A with Computational and Experimental Methods.","authors":"Helge Vatheuer, Oscar Palomino-Hernández, Janis Müller, Phillip Galonska, Serghei Glinca, Paul Czodrowski","doi":"10.1002/cmdc.202400953","DOIUrl":"10.1002/cmdc.202400953","url":null,"abstract":"<p><p>Protonation states serve as an essential molecular recognition motif for biological processes. Their correct consideration is key to successful drug design campaigns, since chemoinformatic tools usually deal with default protonation states of ligands and proteins and miss atypical protonation states. The protonation pattern for the Endothiapepsin/PepstatinA (EP/pepA) complex is investigated using different dry lab and wet lab techniques. ITC experiments revealed an uptake of more than one mole of protons upon pepA binding to EP. Since these experiments were performed at physiological conditions (and not at pH=4.6 at which a large variety of crystal structures is available), a novel crystal structure at pH=7.6 was determined. This crystal structure showed that only modest structural changes occur upon increasing the pH value. This lead to computational studies Poisson-Boltzmann calculations and constant pH MD simulation to reveal the exact location of the protonation event. Both computational studies could reveal a significant pKa shift resulting in non-default protonation state and that the catalytic dyad is responsible for the uptake of protons. This study shows that assessing protonation states for two separate systems (protein and ligand) might result in the incorrect assignment of protonation states and hence incorrect calculation of binding energy.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202400953"},"PeriodicalIF":3.6,"publicationDate":"2025-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142977018","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}
Aminoglycoside-phosphotransferases (APHs) are a class of bacterial enzymes that mediate acquired resistance to aminoglycoside antibiotics. Here we report the identification of small molecules counteracting aminoglycoside resistance in Enterococcus casseliflavus. Molecular dynamics simulations were performed to identify an allosteric pocket in three APH enzymes belonging to 3' and 2'' subfamilies in which we then screened, in silico, 12,000 small molecules. From a subset of only 14 high-scored molecules tested in vitro, we identified a compound, named here EK3, able to non-competitively inhibit the APH(2'')-IVa, an enzyme mediating clinical gentamicin resistance. Structure-activity relationship (SAR) exploration of this hit compound allowed us to identify a molecule with improved enzymatic inhibition. By measuring bacterial sensitivity, we found that the three best compounds in this series restored bactericidal activity of various aminoglycosides, including gentamicin, without exhibiting toxicity to HeLa cells. This work not only provides a basis to fight aminoglycoside resistance but also highlights a proof-of-concept for the search of allosteric modulators by using in silico methods.
{"title":"APH Inhibitors that Reverse Aminoglycoside Resistance in Enterococcus casseliflavus.","authors":"Elise Kaplan, Laurent Chaloin, Jean-François Guichou, Kévin Berrou, Rahila Rahimova, Gilles Labesse, Corinne Lionne","doi":"10.1002/cmdc.202400842","DOIUrl":"10.1002/cmdc.202400842","url":null,"abstract":"<p><p>Aminoglycoside-phosphotransferases (APHs) are a class of bacterial enzymes that mediate acquired resistance to aminoglycoside antibiotics. Here we report the identification of small molecules counteracting aminoglycoside resistance in Enterococcus casseliflavus. Molecular dynamics simulations were performed to identify an allosteric pocket in three APH enzymes belonging to 3' and 2'' subfamilies in which we then screened, in silico, 12,000 small molecules. From a subset of only 14 high-scored molecules tested in vitro, we identified a compound, named here EK3, able to non-competitively inhibit the APH(2'')-IVa, an enzyme mediating clinical gentamicin resistance. Structure-activity relationship (SAR) exploration of this hit compound allowed us to identify a molecule with improved enzymatic inhibition. By measuring bacterial sensitivity, we found that the three best compounds in this series restored bactericidal activity of various aminoglycosides, including gentamicin, without exhibiting toxicity to HeLa cells. This work not only provides a basis to fight aminoglycoside resistance but also highlights a proof-of-concept for the search of allosteric modulators by using in silico methods.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202400842"},"PeriodicalIF":3.6,"publicationDate":"2025-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142968990","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}
Daniil Spector, Vladislav Bykusov, Yulia Isaeva, Roman Akasov, Anastasia Zharova, Igor Rodin, Mikhail Vokuev, Yuri Grishin, Vita Nikitina, Alexander Martynov, Vladimir Kuzmin, Elena Beloglazkina, Olga Krasnovskaya
Light induced release of cisplatin from Pt(IV) prodrugs is a promising tool for precise spatiotemporal control over the antiproliferative activity of Pt-based chemotherapeutic drugs. A combination of light-controlled chemotherapy (PACT) and photodynamic therapy (PDT) in one molecule has the potential to overcome crucial drawbacks of both Pt-based chemotherapy and PDT via a synergetic effect. Herein we report green-light-activated Pt(IV) prodrug GreenPt with BODIPY-based photosentitizer in the axial position with an incredible high light response and singlet oxygen generation ability. GreenPt demonstrated the ability to release cisplatin under low-dose green light irradiation up to 1 J/cm2. The investigation of the photoreduction mechanism of GreenPt prodrug using DFT modeling and ΔG0 PET estimation revealed that the anion-radical formation and substituent photoinduced electron transfer from the triplet excited state of the BODIPY axial ligand to the Pt(IV) center is the key step in the light-induced release of cisplatin. Green-light-activated BODIPY-based photosentitizers 5 and 8 demonstrated outstanding photosensitizing properties with an extraordinary phototoxicity index (PI)>1300. GreenPt prodrug demonstrated gradual intracellular accumulation and light-induced phototoxicity with PI>100, thus demonstrating dual action through light-controlled release of both cisplatin and a potent BODIPY-based photosensitizer.
{"title":"Green Light Activated Dual-Action Pt(IV) Prodrug with Enhanced PDT Activity.","authors":"Daniil Spector, Vladislav Bykusov, Yulia Isaeva, Roman Akasov, Anastasia Zharova, Igor Rodin, Mikhail Vokuev, Yuri Grishin, Vita Nikitina, Alexander Martynov, Vladimir Kuzmin, Elena Beloglazkina, Olga Krasnovskaya","doi":"10.1002/cmdc.202400786","DOIUrl":"10.1002/cmdc.202400786","url":null,"abstract":"<p><p>Light induced release of cisplatin from Pt(IV) prodrugs is a promising tool for precise spatiotemporal control over the antiproliferative activity of Pt-based chemotherapeutic drugs. A combination of light-controlled chemotherapy (PACT) and photodynamic therapy (PDT) in one molecule has the potential to overcome crucial drawbacks of both Pt-based chemotherapy and PDT via a synergetic effect. Herein we report green-light-activated Pt(IV) prodrug GreenPt with BODIPY-based photosentitizer in the axial position with an incredible high light response and singlet oxygen generation ability. GreenPt demonstrated the ability to release cisplatin under low-dose green light irradiation up to 1 J/cm<sup>2</sup>. The investigation of the photoreduction mechanism of GreenPt prodrug using DFT modeling and ΔG<sub>0</sub> PET estimation revealed that the anion-radical formation and substituent photoinduced electron transfer from the triplet excited state of the BODIPY axial ligand to the Pt(IV) center is the key step in the light-induced release of cisplatin. Green-light-activated BODIPY-based photosentitizers 5 and 8 demonstrated outstanding photosensitizing properties with an extraordinary phototoxicity index (PI)>1300. GreenPt prodrug demonstrated gradual intracellular accumulation and light-induced phototoxicity with PI>100, thus demonstrating dual action through light-controlled release of both cisplatin and a potent BODIPY-based photosensitizer.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202400786"},"PeriodicalIF":3.6,"publicationDate":"2025-01-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142941629","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}
The development of small molecule-based drugs emerged as a cornerstone of modern drug discovery. Structural activity relationship (SAR) studies in medicinal chemistry are crucial for lead optimization, where a subtle change in the substituent can significantly alter its binding affinity with the biological target. Herein, a highly efficient single-atom substitution (SAS) approach has been developed, where sulfur for oxygen strategy is utilized as a powerful molecular editing technique to identify N-vinyl Indole-thiobarbituric acid (6 a) as a novel small molecule-based scaffold with tunable photophysical and antiproliferative activities. A series of NIR-emitting indole-barbituric/thiobarbituric acid conjugates exhibiting aggregation-induced emission (AIE) were prepared, where the replacement of oxygen for sulfur strategy emerged as a magic bullet. On the evaluation of photophysical properties and chemopreventive efficacies, a significant improvement in the absorption and emission profile, cellular uptake, and antiproliferative activity was noted for sulfur counterparts. From the pool of the molecules, the lead molecule 6 a unveils a 55 nm emission shift, 142-fold increased anticancer profile, and ~4-fold elevated cellular uptake. Furthermore, the colocalization experiment unravels the nuclear localization of 6 a, where it causes severe DNA damage, arrests the cell cycle in the G2/M phase, and leads to the activation of p53-mediated apoptosis. Our experimental findings represent 6 a as a potential lead molecule possessing excellent anticancer potency in the HCT 116 cell line and HCT 116-derived 3D spheroid model.
{"title":"Harnessing the Sulfur-for-Oxygen Shift: A Magic Bullet for Dynamic Photophysical and Anticancer Activities of Indole-Barbituric Acid Construct","authors":"Kartikay Tyagi, Reena Kumari, V. Venkatesh","doi":"10.1002/cmdc.202400849","DOIUrl":"10.1002/cmdc.202400849","url":null,"abstract":"<p>The development of small molecule-based drugs emerged as a cornerstone of modern drug discovery. Structural activity relationship (SAR) studies in medicinal chemistry are crucial for lead optimization, where a subtle change in the substituent can significantly alter its binding affinity with the biological target. Herein, a highly efficient single-atom substitution (SAS) approach has been developed, where sulfur for oxygen strategy is utilized as a powerful molecular editing technique to identify N-vinyl Indole-thiobarbituric acid (<b>6 a</b>) as a novel small molecule-based scaffold with tunable photophysical and antiproliferative activities. A series of NIR-emitting indole-barbituric/thiobarbituric acid conjugates exhibiting aggregation-induced emission (AIE) were prepared, where the replacement of oxygen for sulfur strategy emerged as a magic bullet. On the evaluation of photophysical properties and chemopreventive efficacies, a significant improvement in the absorption and emission profile, cellular uptake, and antiproliferative activity was noted for sulfur counterparts. From the pool of the molecules, the lead molecule <b>6 a</b> unveils a 55 nm emission shift, 142-fold increased anticancer profile, and ~4-fold elevated cellular uptake. Furthermore, the colocalization experiment unravels the nuclear localization of <b>6 a</b>, where it causes severe DNA damage, arrests the cell cycle in the G2/M phase, and leads to the activation of p53-mediated apoptosis. Our experimental findings represent <b>6 a</b> as a potential lead molecule possessing excellent anticancer potency in the HCT 116 cell line and HCT 116-derived 3D spheroid model.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":"20 7","pages":""},"PeriodicalIF":3.6,"publicationDate":"2025-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142941661","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}
Dr. Raghuvir H. Gaonkar, Dr. Thibaud Bailly, Dr. Jacopo Millul, Dr. Rosalba Mansi, Dr. Mirja Harms, Prof. Dr. Jan Münch, Prof. Dr. Melpomeni Fani
The C−X-C chemokine receptor 4 (CXCR4) is highly upregulated in most cancers, making it an ideal target for delivering radiation therapy to tumors. We previously demonstrated the feasibility of targeting CXCR4 in vivo using a radiolabeled derivative of EPI-X4, an endogenous CXCR4 antagonist, named DOTA−K-JM#173. However, this derivative showed undesirable accumulation in the kidneys, which would limit its clinical use. In this study, we identified that removing a positive charge from the peptide sequence significantly reduced renal uptake. We evaluated a series of optimized derivatives lacking this positive charge, in vitro and in vivo in a xenografted athymic nude mice model, after radiolabeling with 177Lu. The most promising derivatives were further assessed in vivo after 68Ga labeling. Among them, we identified DOTA-JM#173 and D-L1-DOTA-JM#173, where the D-Ile1 was replaced by D-Leu1, two optimized derivatives with a lysine residue removed. These two molecules represent the most advanced DOTA-conjugated ligands derived from EPI-X4 for CXCR4-directed theranostic applications, offering enhanced potential for targeted cancer treatment.
{"title":"Improving Affinity while Reducing Kidney Uptake of CXCR4-Targeting Radioligands Derived from the Endogenous Antagonist EPI-X4","authors":"Dr. Raghuvir H. Gaonkar, Dr. Thibaud Bailly, Dr. Jacopo Millul, Dr. Rosalba Mansi, Dr. Mirja Harms, Prof. Dr. Jan Münch, Prof. Dr. Melpomeni Fani","doi":"10.1002/cmdc.202400773","DOIUrl":"10.1002/cmdc.202400773","url":null,"abstract":"<p>The C−X-C chemokine receptor 4 (CXCR4) is highly upregulated in most cancers, making it an ideal target for delivering radiation therapy to tumors. We previously demonstrated the feasibility of targeting CXCR4 <i>in vivo</i> using a radiolabeled derivative of EPI-X4, an endogenous CXCR4 antagonist, named DOTA−K-JM#173. However, this derivative showed undesirable accumulation in the kidneys, which would limit its clinical use. In this study, we identified that removing a positive charge from the peptide sequence significantly reduced renal uptake. We evaluated a series of optimized derivatives lacking this positive charge, <i>in vitro</i> and <i>in vivo</i> in a xenografted athymic nude mice model, after radiolabeling with <sup>177</sup>Lu. The most promising derivatives were further assessed <i>in vivo</i> after <sup>68</sup>Ga labeling. Among them, we identified DOTA-JM#173 and D-L<sup>1</sup>-DOTA-JM#173, where the D-Ile<sup>1</sup> was replaced by D-Leu<sup>1</sup>, two optimized derivatives with a lysine residue removed. These two molecules represent the most advanced DOTA-conjugated ligands derived from EPI-X4 for CXCR4-directed theranostic applications, offering enhanced potential for targeted cancer treatment.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":"20 6","pages":""},"PeriodicalIF":3.6,"publicationDate":"2025-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142941705","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}
Teresa Glomb, Dr. Kamila Środa-Pomianek, Dr. Anna Palko-Łabuz, Dr. Olga Wesołowska, Dr. Agnieszka Wikiera, Dr. Kamil Wojtkowiak, Dr. Aneta Jezierska, Dr. Andrzej Kochel, Prof. Roman Lesyk, Dr. Piotr Świątek
In this research, a series of novel hydrazone derivatives based on pyrazolopyridothiazinylacetohydrazide were designed, synthesized, and evaluated for their in vitro cytotoxic potency on several human colon cancer cells (HTC116, HT-29, and LoVo). After MTT and SRB assays four of the most active derivatives: hydrazide GH and hydrazones GH7, GH8, and GH11, were chosen for further investigation. Hydrazone GH11 had the highest cytotoxic activity (IC50 values of c.a. 0.5 μM). Additionally, the impact of novel derivatives on the oxidative stress level, apoptosis induction, and modulation of inflammation in colon cancer cells was examined. In all studies, the activity of the derivatives increased in order GH < GH7 < GH8 < GH11. At the same time, most of the research was conducted on compounds combined with apple pectin (PC). The most interesting observation was that all the studied derivatives applied together with PC showed significantly higher activity than observed in the case of using PC, hydrazide, or hydrazones separately. Finally, computational chemistry methods (molecular modeling and Density Functional Theory – DFT) were used to complement the experimental studies.
{"title":"New Hydrazone Derivatives Based on Pyrazolopyridothiazine Core as Cytotoxic Agents to Colon Cancers: Design, Synthesis, Biological Evaluation, and Molecular Modeling","authors":"Teresa Glomb, Dr. Kamila Środa-Pomianek, Dr. Anna Palko-Łabuz, Dr. Olga Wesołowska, Dr. Agnieszka Wikiera, Dr. Kamil Wojtkowiak, Dr. Aneta Jezierska, Dr. Andrzej Kochel, Prof. Roman Lesyk, Dr. Piotr Świątek","doi":"10.1002/cmdc.202400687","DOIUrl":"10.1002/cmdc.202400687","url":null,"abstract":"<p>In this research, a series of novel hydrazone derivatives based on pyrazolopyridothiazinylacetohydrazide were designed, synthesized, and evaluated for their <i>in vitro</i> cytotoxic potency on several human colon cancer cells (HTC116, HT-29, and LoVo). After MTT and SRB assays four of the most active derivatives: hydrazide <b>GH</b> and hydrazones <b>GH7</b>, <b>GH8</b>, and <b>GH11</b>, were chosen for further investigation. Hydrazone <b>GH11</b> had the highest cytotoxic activity (IC<sub>50</sub> values of c.a. 0.5 μM). Additionally, the impact of novel derivatives on the oxidative stress level, apoptosis induction, and modulation of inflammation in colon cancer cells was examined. In all studies, the activity of the derivatives increased in order <b>GH</b> < <b>GH7</b> < <b>GH8</b> < <b>GH11</b>. At the same time, most of the research was conducted on compounds combined with apple pectin (PC). The most interesting observation was that all the studied derivatives applied together with PC showed significantly higher activity than observed in the case of using PC, hydrazide, or hydrazones separately. Finally, computational chemistry methods (molecular modeling and Density Functional Theory – DFT) were used to complement the experimental studies.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":"20 7","pages":""},"PeriodicalIF":3.6,"publicationDate":"2025-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142941711","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}
Dr. Alexandre Specht, Maxime Klimezak, Dr. Sidney Cambridge
New concepts to treat eye diseases have emerged that elegantly combine unnatural light exposure with chemical biology approaches to achieve superior cellular specificity and, as a result, improvement of visual function. Historically, light exposure without further molecular eye treatment has offered limited success including photocoagulation to halt pathological blood vessel growth or low light exposure to stimulate retinal cell viability. To add cellular specificity to such treatments, researchers have introduced various biological or chemical light-sensing molecules and combined those with light exposure. (Pre-)clinical trials describe the use of optogenetics and channelrhodpsins, i. e. light-sensitive ion channels, in patient vision restoration. In the chemical arena, pharmacological agents, rendered light-sensitive by reversible modification with photosensitive protecting compounds (“caging”), have been applied to eyes of living mice to photo-release specific cellular activities. Among these were successful proof-of-principle experiments that were conducted to establish photo-sensitive gene therapies in the eye. For light-mediated treatment in combination with chemical biology, we wish to describe here the current frontiers of research in vision restoration with an eye on differences between biological and chemical light-sensing molecules, patient requirements, and future outlooks.
{"title":"Seeing in the Future – a Perspective on Combining Light with Chemical Biology Approaches to Treat Retinal Pathologies","authors":"Dr. Alexandre Specht, Maxime Klimezak, Dr. Sidney Cambridge","doi":"10.1002/cmdc.202400827","DOIUrl":"10.1002/cmdc.202400827","url":null,"abstract":"<p>New concepts to treat eye diseases have emerged that elegantly combine unnatural light exposure with chemical biology approaches to achieve superior cellular specificity and, as a result, improvement of visual function. Historically, light exposure without further molecular eye treatment has offered limited success including photocoagulation to halt pathological blood vessel growth or low light exposure to stimulate retinal cell viability. To add cellular specificity to such treatments, researchers have introduced various biological or chemical light-sensing molecules and combined those with light exposure. (Pre-)clinical trials describe the use of optogenetics and channelrhodpsins, i. e. light-sensitive ion channels, in patient vision restoration. In the chemical arena, pharmacological agents, rendered light-sensitive by reversible modification with photosensitive protecting compounds (“caging”), have been applied to eyes of living mice to photo-release specific cellular activities. Among these were successful proof-of-principle experiments that were conducted to establish photo-sensitive gene therapies in the eye. For light-mediated treatment in combination with chemical biology, we wish to describe here the current frontiers of research in vision restoration with an eye on differences between biological and chemical light-sensing molecules, patient requirements, and future outlooks.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":"20 7","pages":""},"PeriodicalIF":3.6,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142941807","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}
Gemcitabine (GEM), a chemotherapeutic agent, is widely used to treat various neoplastic conditions, such as pancreatic, lung, breast, and ovarian cancer. However, its therapeutic effectiveness is often hindered by its short half-life and susceptibility to enzymatic degradation. To address these limitations, in this research, five new conjugates of GEM were synthesized by conjugating its N-4 amino group with five different acids [4-decenoic acid (4Dec), lipoic acid (Lipo), lauric acid (Laur), 5-benzyl N-(tert-butoxycarbonyl)- L-glutamate (Glu), and decanoic acid (Dec)]. The anticancer potential of these conjugates was evaluated using CCK-8 assay. Among the synthesized conjugates, 4Dec-GEM demonstrated comparable cytotoxic activity to native GEM. The mechanistic insight of 4Dec-GEM was investigated using annexin-V FITC/propidium iodide staining, reactive oxygen species generation, and mitochondrial membrane potential loss assays. To further enhance its therapeutic efficacy, 4Dec-GEM was encapsulated into poly(lactic-co-glycolic acid) (PLGA) nanoparticles using single-emulsion method using high-pressure homogenization. The developed nanoparticles were characterized by various techniques (TEM, FT-IR, DSC, p-XRD) and demonstrated successful entrapment of 4Dec-GEM inside PLGA nanoparticles. Finally, the cytotoxicity of developed nanoparticles demonstrated improved anticancer activity as compared to native GEM in cancerous cell lines. Our study demonstrated that the combination of prodrug and nanoparticle approach can be a promising approach to augment the therapeutic efficacy of GEM.
吉西他滨(GEM)是一种化疗药物,广泛用于治疗各种肿瘤,如胰腺癌、肺癌、乳腺癌和卵巢癌。然而,其治疗效果往往受到其亲水性,半衰期短和酶降解敏感性的阻碍。为了解决这些局限性,本研究通过将GEM的N-4氨基与5种不同的酸[4-癸烯酸(4Dec)、硫辛酸(Lipo)、月桂酸(Laur)、5-苄基N-(叔丁基羰基)- l -谷氨酸(Glu)和癸酸(Dec)]偶联,合成了5种新的GEM前药。通过CCK-8、膜联蛋白- v FITC/碘化丙啶染色、ROS和线粒体膜电位损失测定来评估这些前药的抗癌潜力。在这些偶联物中,4Dec-GEM表现出与天然GEM相当的细胞毒活性。为了进一步提高其治疗效果,利用前药的亲脂性,利用单乳和高压均质技术将4Dec-GEM包裹在聚乳酸-羟基乙酸(PLGA)纳米颗粒中。通过各种技术对所制备的纳米颗粒进行了表征,并成功地将4Dec-GEM包埋在PLGA纳米颗粒中。最后,在A549、MIA-PaCa-2和PANC-1癌细胞系中,与天然GEM相比,开发的纳米颗粒的细胞毒性显示出更好的治疗效果。我们的研究表明,前体药物和纳米颗粒的结合是提高GEM治疗效果的一种很有前景的方法。
{"title":"Synthesis of a Gemcitabine Prodrug and its Encapsulation into Polymeric Nanoparticles for Improved Therapeutic Efficacy","authors":"Kajal Kaliya, Neha Bhardwaj, Ruchika, Ankit Saneja","doi":"10.1002/cmdc.202400532","DOIUrl":"10.1002/cmdc.202400532","url":null,"abstract":"<p>Gemcitabine (GEM), a chemotherapeutic agent, is widely used to treat various neoplastic conditions, such as pancreatic, lung, breast, and ovarian cancer. However, its therapeutic effectiveness is often hindered by its short half-life and susceptibility to enzymatic degradation. To address these limitations, in this research, five new conjugates of GEM were synthesized by conjugating its N-4 amino group with five different acids [4-decenoic acid (4Dec), lipoic acid (Lipo), lauric acid (Laur), 5-benzyl N-(tert-butoxycarbonyl)- L-glutamate (Glu), and decanoic acid (Dec)]. The anticancer potential of these conjugates was evaluated using CCK-8 assay. Among the synthesized conjugates, 4Dec-GEM demonstrated comparable cytotoxic activity to native GEM. The mechanistic insight of 4Dec-GEM was investigated using annexin-V FITC/propidium iodide staining, reactive oxygen species generation, and mitochondrial membrane potential loss assays. To further enhance its therapeutic efficacy, 4Dec-GEM was encapsulated into poly(lactic-co-glycolic acid) (PLGA) nanoparticles using single-emulsion method using high-pressure homogenization. The developed nanoparticles were characterized by various techniques (TEM, FT-IR, DSC, p-XRD) and demonstrated successful entrapment of 4Dec-GEM inside PLGA nanoparticles. Finally, the cytotoxicity of developed nanoparticles demonstrated improved anticancer activity as compared to native GEM in cancerous cell lines. Our study demonstrated that the combination of prodrug and nanoparticle approach can be a promising approach to augment the therapeutic efficacy of GEM.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":"20 7","pages":""},"PeriodicalIF":3.6,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142941808","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}
Correction to “α-Triazolylboronic Acids: A Novel Scaffold to Target FLT3 in AML”
Maria Luisa Introvigne, Lorenza Destro, Luca Mologni, Valentina Crippa, Paolo Zardi, Francesco Fini, Fabio Prati, Emilia Caselli, Alfonso Zambon, “α-Triazolylboronic Acids: A Novel Scaffold to Target FLT3 in AML”, ChemMedChem2024, 19, e202400622 DOI: https://doi.org/10.1002/cmdc.202400622
The full first names of the authors of this paper were omitted upon initial publication, and Fabio Prati was incorrectly entered as “L. F. Prati”. All author names are correct and complete as shown.
{"title":"CORRIGENDUM: α-Triazolylboronic Acids: A Novel Scaffold to Target FLT3 in AML","authors":"","doi":"10.1002/cmdc.202400982","DOIUrl":"10.1002/cmdc.202400982","url":null,"abstract":"<p>Correction to “α-Triazolylboronic Acids: A Novel Scaffold to Target FLT3 in AML”</p><p>Maria Luisa Introvigne, Lorenza Destro, Luca Mologni, Valentina Crippa, Paolo Zardi, Francesco Fini, Fabio Prati, Emilia Caselli, Alfonso Zambon, “α-Triazolylboronic Acids: A Novel Scaffold to Target FLT3 in AML”, <i>ChemMedChem</i> <b>2024</b>, <i>19</i>, e202400622 DOI: https://doi.org/10.1002/cmdc.202400622</p><p>The full first names of the authors of this paper were omitted upon initial publication, and Fabio Prati was incorrectly entered as “L. F. Prati”. All author names are correct and complete as shown.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":"20 6","pages":""},"PeriodicalIF":3.6,"publicationDate":"2025-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cmdc.202400982","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142930143","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}