19,578 results on '"Alkylating agents"'
Search Results
2. Safety and Preliminary Effectiveness of BNT327, an Investigational Therapy for Breast Cancer, When Given in Combination With Chemotherapy
- Published
- 2024
3. The synergy between alkylating agents and ERCC1–XPF inhibitors is p53 dependent.
- Author
-
Ciniero, Gloria, Pedro, Tiago Marques, Dumontet, Charles, Elmenoufy, Ahmed H., West, Frederick G., Weinfeld, Michael, Gentile, Francesco, Tuszynski, Jack A., Cros‐Perrial, Emeline, and Jordheim, Lars Petter
- Subjects
- *
EXCISION repair , *CELL transformation , *ALKYLATING agents , *CELL determination , *CELL survival , *DNA repair - Abstract
Background Objectives Methods Results Conclusion DNA repair plays a major role in maintaining genomic stability, thus limiting the transformation of normal cells into cancer cells. However, in cancer patients treated with DNA‐targeting drugs, DNA repair can decrease efficacy by removing the damage generated by such molecules that is needed to induce pharmacological activity. Inhibiting DNA repair thus represents an interesting approach to potentiating the activity of chemotherapy in this setting.Here, we continue the characterization of an inhibitor of the interaction between Excision Repair Cross‐Complementing Rrodent repair deficiency complementation group 1 (ERCC1) and Xeroderma Pigmentousum group F (XPF) (
B9 ), two key proteins of nucleotide excision repair.We used various cell lines and co‐incubation studies for the determination of cell survival and DNA repair capacities.We show that it is synergistic with other platinum derivatives than previously described, and that synergy is lacking in cells not expressing ERCC1 or XPF. Finally, a series of experiments show that potentiation is observed only in cells expressing wild‐type p53.Our results confirm the mechanism of action of our ERCC1–XPF inhibitor and give important additional data on this approach to enhance the activity of already existing cancer drugs. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
4. The third generation AKR1C3-activated prodrug, ACHM-025, eradicates disease in preclinical models of aggressive T-cell acute lymphoblastic leukemia.
- Author
-
Toscan, Cara E., McCalmont, Hannah, Ashoorzadeh, Amir, Lin, Xiaojing, Fu, Zhe, Doculara, Louise, Kosasih, Hansen J., Cadiz, Roxanne, Zhou, Anthony, Williams, Sarah, Evans, Kathryn, Khalili, Faezeh, Cai, Ruilin, Yeats, Kristy L., Gifford, Andrew J., Pickford, Russell, Mayoh, Chelsea, Xie, Jinhan, Henderson, Michelle J., and Trahair, Toby N.
- Subjects
LYMPHOBLASTIC leukemia ,HEMATOLOGIC malignancies ,ALKYLATING agents ,NITROGEN mustards ,ACUTE leukemia - Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy that expresses high levels of the enzyme aldo-keto reductase family 1 member C3 (AKR1C3). To exploit this finding, we developed a novel prodrug, ACHM-025, which is selectively activated by AKR1C3 to a nitrogen mustard DNA alkylating agent. We show that ACHM-025 has potent in vivo efficacy against T-ALL patient-derived xenografts (PDXs) and eradicated the disease in 7 PDXs. ACHM-025 was significantly more effective than cyclophosphamide both as a single agent and when used in combination with cytarabine/6-mercaptopurine. Notably, ACHM-025 in combination with nelarabine was curative when used to treat a chemoresistant T-ALL PDX in vivo. The in vivo efficacy of ACHM-025 directly correlated with AKR1C3 expression levels, providing a predictive biomarker for response. Together, our work provides strong preclinical evidence highlighting the potential of ACHM-025 as a targeted and effective therapy for aggressive forms of T-ALL. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. Synergistic Combination of Quercetin and Mafosfamide in Treatment of Bladder Cancer Cells.
- Author
-
Spagnuolo, Carmela, Mautone, Francesco, Meola, Anna Maria Iole, Moccia, Stefania, Di Lorenzo, Giuseppe, Buonerba, Carlo, and Russo, Gian Luigi
- Subjects
- *
TRANSITIONAL cell carcinoma , *ALKYLATING agents , *CELL cycle , *BLADDER cancer , *QUERCETIN - Abstract
Bladder cancer, which has a rising incidence, is the 10th most common cancer. The transitional cell carcinoma histotype is aggressive and often current therapies are ineffective. We investigated the anti-proliferative effect of quercetin, a natural flavonoid, in combination with the alkylating agent mafosfamide (MFA) on two human bladder cancer cell lines, namely RT112 and J82, representing the progression from low-grade to high-grade tumors, respectively. In both cell types, the combined treatment led to a synergic reduction in cell viability confirmed by a combination index of less than one, though different biological responses were noted. In J82 cells, MFA alone and, to a lesser extent, with quercetin caused cell cycle arrest in the G2/M phase, but only the combined treatment triggered apoptotic cell death. In contrast, in RT112 cells, quercetin induced autophagy, evidenced by the autophagosome formation and the increase in LC-3 lipidation. Interestingly, the synergistic effect was observed only when cells were pre-treated with MFA for 24 h before adding quercetin, not in the reverse order. This suggests that quercetin may help overcome MFA resistance to apoptosis. Although further studies are needed, investigating the combined effects of quercetin and MFA could help elucidate the mechanisms of drug resistance in bladder cancer treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Synthesis of New Pyrazolo[3,4- d ]pyrimidine Derivatives: NMR Spectroscopic Characterization, X-Ray, Hirshfeld Surface Analysis, DFT, Molecular Docking, and Antiproliferative Activity Investigations.
- Author
-
El Hafi, Mohamed, Anouar, El Hassane, Lahmidi, Sanae, Boulhaoua, Mohammed, Loubidi, Mohammed, Alanazi, Ashwag S., Filali, Insaf, Hefnawy, Mohamed, El Ghayati, Lhoussaine, Mague, Joel T., and Essassi, El Mokhtar
- Subjects
- *
MOLECULAR structure , *PROPARGYL bromide , *SURFACE analysis , *ALKYLATING agents , *MOLECULAR docking , *PYRIMIDINES - Abstract
Four new pyrazolo[3,4-d]pyrimidines (P1–P4) were successfully synthesized in good relative yields by reacting 3-methyl-1-phenyl-1H-pyrazolo[3,4-d]pyrimidin-4-ol with various alkylating agents (methyl iodide, propargyl bromide, and phenacyl bromide) at room temperature in DMF solvent, employing liquid–solid phase transfer catalysis. The P1–P4 structures were elucidated using NMR spectroscopy and X-ray diffraction. Intermolecular interactions in P1–P4 were analyzed via Hirshfeld surface analysis and 2D fingerprint plots. Geometrical parameters were accurately modeled by DFT calculations using the B3LYP hybrid functional combined with a 6–311++G(d,p) basis set. The antiproliferative activity of P1–P4 towards colorectal carcinoma (HCT 116), human hepatocellular carcinoma (HepG2), and human breast cancer (MCF-7) cell lines, along with one normal cell line (WI38) was investigated using the MTT assay and sunitinib as a reference. Compounds P1 and P2 exhibited antiproliferative activities comparable to the reference drug towards all tested cells, with an IC50 range of 22.7–40.75 µM. Both compounds also showed high selectivity indices and minimal cytotoxic effects on the normal cell line. Molecular docking revealed that the significant antiproliferative activity may attributed to the number and type of intermolecular hydrogen bonding established between pyrazolo[3,4-d]pyrimidines and DNA topoisomerase, a common target for various anticancer agents. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Moderate India Pale Ale beer consumption promotes antigenotoxic and non‐mutagenic effects in ex vivo and in vivo mice models.
- Author
-
de Cordova Kindermann, Schellen, Caon, Glauco, Boeck, Carina Rodrigues, de Oliveira Bauer, Carla, dos Santos da Silva, Nicollas, Possamai, Otavio Lucio, Longaretti, Luiza Martins, Magenis, Marina Lummertz, Damiani, Adriani Paganini, de Oliveira Monteiro, Isadora, and de Andrade, Vanessa Moraes
- Subjects
- *
INDIA pale ale , *CRAFT beer , *ALCOHOL drinking , *ALKYLATING agents , *BLOOD collection - Abstract
BACKGROUND: Discussion of the benefits of moderate alcohol consumption is ongoing. Broadly, research focusing on ethanol consumption tends to report no benefits. However, studies that distinguish between different types of alcoholic beverages, particularly beers, often reveal positive effects. The present study evaluated the genotoxic and mutagenic effects of moderate chronic consumption of India Pale Ale (IPA) craft beer. Sixty‐four adult male Swiss mice were used and divided into control and treatment groups receiving water, IPA beer with 55.23 g of ethanol per liter of beer, aqueous solution with 55.23 g of ethanol per liter, and hop infusion ad libitum for 30 days. After this period, the animals were genetically evaluated with a comet assay. For the ex vivo comet assay, blood was collected and exposed to hydrogen peroxide (H2O2). For the in vivo assay, the alkylating agent cyclophosphamide (CP) was administered to the groups after blood collection and sacrificed after 24 h. Brain, liver, and heart tissues were analyzed. Bone marrow was collected and submitted to the micronucleus test. RESULTS: The groups treated with IPA beer, ethanol, and hops did not show genotoxic and mutagenic action in the blood, brain, heart, or liver. The antigenotoxic action of IPA beer and hops was observed in both in vivo and ex vivo models, showing a similar reduction in DNA damage caused by CP. There was no significant difference between the groups with regard to the formation of micronuclei by CP. CONCLUSION: Moderate chronic consumption of IPA beer and hops infusion showed antigenotoxic effects in mice but no antimutagenic action. © 2024 Society of Chemical Industry. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. Prophylactic Antimicrobials for Prevention of Febrile Neutropenia in Tumour‐Bearing Dogs Treated With Lomustine.
- Author
-
Gumash, Meredith, Martin, Olya A., Lindley, Stephanie E. S., and Zhu, Xiaojuan
- Subjects
- *
BODY surface area , *FEBRILE neutropenia , *ALKYLATING agents , *BODY weight , *DOGS - Abstract
ABSTRACT CCNU (1‐[2‐chloroethyl]‐3‐cyclohexyl‐1‐nitrosurea), lomustine, is an oral alkylating agent in the nitrosourea subgroup. The dose‐limiting toxicity of CCNU is neutropenia most frequently documented 7 days after its administration. Use of prophylactic antimicrobials to prevent chemotherapy‐related febrile neutropenia (FN) and its associated morbidity and mortality has been well‐documented in human oncology, but this information is limited in the veterinary literature. The purpose of this multi‐institutional retrospective study was to assess whether antimicrobial prophylaxis reduced the risk of FN approximately 7 days after CCNU administration in tumour‐bearing dogs. A secondary goal was to identify risk factors for fever development in neutropenic dogs. Two hundred dogs were included in the study. One hundred and fifty‐three dogs (76.5%) were neutropenic at the first post‐CCNU recheck. One hundred and six (69.3%) dogs received prophylactic antimicrobials and 47 (30.7%) did not. Of the 106 dogs on prophylactic antimicrobials, 8 (7.5%) developed FN. Of the 47 dogs in the no‐prophylactic antimicrobials group, 4 (8.5%) developed FN. Use of prophylactic antimicrobials did not reduce the risk of development of FN (p = 0.84). Older age (> 9 y), lower weight and body surface area, and pre‐treatment with chemotherapy or radiation therapy were significantly associated with development of FN (p = 0.009, p = 0.023, p = 0.015 and p = 0.01). Patients with a lower absolute neutrophil count, and a higher VCOG‐CTCAE v2 neutropenia grade were also at an increased risk of developing FN (p = 0.01, p < 0.001). Additional studies may help establish guidelines for antimicrobial prophylaxis in dogs treated with CCNU, especially for those at high‐risk for FN. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Longitudinal trends in testicular volume z scores from puberty to adulthood, sperm quality, and paternity outcomes after childhood cancer.
- Author
-
Korhonen, Melanie, Jahnukainen, Kirsi, and Koskela, Mikael
- Subjects
- *
EXPOSURE therapy , *ALKYLATING agents , *CHILDREN'S hospitals , *AZOOSPERMIA , *CHILDHOOD cancer - Abstract
ABSTRACT Background Methods Results Conclusions Childhood cancer therapy may cause subfertility. This study correlated cancer therapy exposures with testicular volumes from puberty to adulthood, spermatogenesis, and paternity outcomes in adulthood.The study population comprised 255 male childhood cancer survivors (CCS) (survival ≥5 years, diagnosed in 1964–2000 at the Helsinki Children's Hospital) whose testicular volume was measured at ages 12 years (
n = 38), 14 years (n = 57), 16 years (n = 63), 18 years (n = 105), and in adulthood (n = 43; median age, 27 years). Testicular volumes were converted to age‐specific z scores. In addition, 92 CCS provided semen sample in adulthood (median age, 25.2 years); and paternity was evaluated through national register data (mean age at assessment, 37.6 years;n = 252).Compared with age‐specific reference values, CCS generally exhibited low testicular volume z scores at ages 12–18 years. Testicular volume z scores in CCS treated exclusively with chemotherapy returned to the reference range in adulthood. In contrast, patients exposed to testicular radiation ≥1 gray (Gy) (median dose, 12 Gy) showed no late recovery in testicular size. Testicular radiation ≥1 Gy and a cyclophosphamide equivalent dose ≥12 g/m2 were identified as risk factors for azoospermia in adulthood. Patients exposed to testicular radiation ≥1 Gy and a cyclophosphamide equivalent dose ≥4 g/m2 had lower paternity rates.Testicular volume growth after prolonged follow‐up suggests a potential late recovery of spermatogenesis in CCS treated exclusively with chemotherapy. However, alkylating agents increased the risk of having prolonged azoospermia and nonpaternity. High‐dose testicular radiation causes long‐term depletion of spermatogonia and was the strongest risk factor for azoospermia and nonpaternity. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
10. Engineered 1,6-heptadiynes based polymeric prodrug system for anticancer drug delivery.
- Author
-
Kumar, Pawan, Patra, Diptendu, Halder, Ratan, and Shunmugam, Raja
- Subjects
- *
DRUG delivery systems , *TRANSMISSION electron microscopes , *ALKYLATING agents , *ETHYLENE glycol , *LIGHT transmission - Abstract
AbstractDiacetylene moiety,
DIOL was functionalized to chemotherapeutic drug, chlorambucil and water-soluble moiety poly(ethylene glycol) to get our macromonomerMPDC .MPDC was further cyclopolymerized using Hoveyda Grubbs’ second generation catalyst, results to form polymerPMPDC . Amphiphilic nature ofPMPDC leads to self-assembly in water which was characterized by dynamic light scattering and transmission electron microscope. Cytotoxicity study was performed with HeLa and MCF 7 cancer cell lines which showed good cell growth inhibition. AsPMPDC having non-fluorescent property, another polymerPMPDP was synthesized as control using 1-pyrenebutyric acid which was further used for drug release at different pH and cell internalization study. Increase in internalization of polymer with increase in concentration was observed in both HeLa and MCF 7 cells. Formation of all the compounds were characterized carefully by different analytical techniques. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
11. S-allyl-cysteine triggers cytotoxic events in rat glioblastoma RG2 and C6 cells and improves the effect of temozolomide through the regulation of oxidative responses.
- Author
-
Reyes-Soto, Carolina Y., Ramírez-Carreto, Ricardo J., Ortíz-Alegría, Luz Belinda, Silva-Palacios, Alejandro, Zazueta, Cecilia, Galván-Arzate, Sonia, Karasu, Çimen, Túnez, Isaac, Tinkov, Alexey A., Aschner, Michael, López-Goerne, Tessy, Chavarría, Anahí, and Santamaría, Abel
- Subjects
CENTRAL nervous system cancer ,ALKYLATING agents ,REACTIVE oxygen species ,CELL lines ,TEMOZOLOMIDE - Abstract
Glioblastoma (GBM) is an aggressive form of cancer affecting the Central Nervous System (CNS) of thousands of people every year. Redox alterations have been shown to play a key role in the development and progression of these tumors as Reactive Oxygen Species (ROS) formation is involved in the modulation of several signaling pathways, transcription factors, and cytokine formation. The second-generation oral alkylating agent temozolomide (TMZ) is the first-line chemotherapeutic drug used to treat of GBM, though patients often develop primary and secondary resistance, reducing its efficacy. Antioxidants represent promising and potential coadjutant agents as they can reduce excessive ROS formation derived from chemo- and radiotherapy, while decreasing pharmacological resistance. S-allyl-cysteine (SAC) has been shown to inhibit the proliferation of several types of cancer cells, though its precise antiproliferative mechanisms remain poorly investigated. To date, SAC effects have been poorly explored in GBM cells. Here, we investigated the effects of SAC in vitro, either alone or in combination with TMZ, on several toxic and modulatory endpoints—including oxidative stress markers and transcriptional regulation—in two glioblastoma cell lines from rats, RG2 and C6, to elucidate some of the biochemical and cellular mechanisms underlying its antiproliferative properties. SAC (1–750 µM) decreased cell viability in both cell lines in a concentration-dependent manner, although C6 cells were more resistant to SAC at several of the tested concentrations. TMZ also produced a concentration-dependent effect, decreasing cell viability of both cell lines. In combination, SAC (1 µM or 100 µM) and TMZ (500 µM) enhanced the effects of each other. SAC also augmented the lipoperoxidative effect of TMZ and reduced cell antioxidant resistance in both cell lines by decreasing the TMZ-induced increase in the GSH/GSSG ratio. In RG2 and C6 cells, SAC per se had no effect on Nrf2/ARE binding activity, while in RG2 cells TMZ and the combination of SAC + TMZ decreased this activity. Our results demonstrate that SAC, alone or in combination with TMZ, exerts antitumor effects mediated by regulatory mechanisms of redox activity responses. SAC is also a safe drug for testing in other models as it produces non-toxic effects in primary astrocytes. Combined, these effects suggest that SAC affords antioxidant properties and potential antitumor efficacy against GBM. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. Glioblastoma Sensitization to Therapeutic Effects by Glutamine Deprivation Depends on Cellular Phenotype and Metabolism.
- Author
-
Isakova, Alina A., Druzhkova, Irina N., Mozherov, Artem M., Mazur, Diana V., Antipova, Nadezhda V., Krasnov, Kirill S., Fadeev, Roman S., Gasparian, Marine E., and Yagolovich, Anne V.
- Subjects
- *
ALKYLATING agents , *TREATMENT effectiveness , *AMINO acids , *CELL analysis , *GLUTAMINE , *OXIDATIVE phosphorylation - Abstract
Glutamine plays an important role in tumor metabolism. It is known that the core region of solid tumors is deprived of glutamine, which affects tumor growth and spread. Here we investigated the effect of glutamine deprivation on cellular metabolism and sensitivity of human glioblastoma cells U87MG and T98G to drugs of various origin: alkylating cytostatic agent temozolomide; cytokine TRAIL DR5-B – agonist of the DR5 receptor; and GMX1778 – a targeted inhibitor of the enzyme nicotinamide phosphoribosyltransferase (NAMPT), limiting NAD biosynthesis. Bioinformatics analysis of the cell transcriptomes showed that U87MG cells have a more differentiated phenotype than T98G, and also differ in the expression profile of the genes associated with glutamine metabolism. Upon glutamine deprivation, growth rate of the U87MG and T98G cells decreased. Analysis of cellular metabolism by FLIM microscopy of NADH as well as assessment of lactate content in the medium showed that glutamine deprivation shifted metabolic status of the U87MG cells towards glycolysis. This was accompanied by the increase in expression of the stemness marker CD133, which collectively could indicate de-differentiation of these cells. At the same time, we observed increase in both expression of the DR5 receptor and sensitivity of the U87MG cells to DR5-B. On the contrary, glutamine deprivation of T98G cells induced metabolic shift towards oxidative phosphorylation, decrease in the DR5 expression and resistance to DR5-B. The effects of NAMPT inhibition also differed between the two cell lines and were opposite to the effects of DR5-B: upon glutamine deprivation, U87MG cells acquired resistance, while T98G cells were sensitized to GMX1778. Thus, phenotypic and metabolic differences between the two human glioblastoma cell lines caused divergent metabolic changes and contrasting responses to different targeted drugs during glutamine deprivation. These data should be considered when developing treatment strategies for glioblastoma via drug-mediated deprivation of amino acids, as well as when exploring novel therapeutic targets. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
13. Supported Nickel Nanoparticles as Catalyst in Direct sp3 C−H Alkylation of 9H‐Fluorene Using Alcohols as Alkylating Agent.
- Author
-
M, Vageesh, Joshi, Harsh, A S, Anupriya, and Dey, Raju
- Subjects
- *
ALKYLATING agents , *HETEROGENEOUS catalysis , *NICKEL catalysts , *TRANSITION metals , *ALKYLATION - Abstract
Herein, we report an inexpensive first‐row transition metal Ni heterogeneous catalytic system for the Csp3‐mono alkylation of fluorene using alcohols as alkylating agents
via borrowing hydrogen strategy. The catalytic protocol displayed versatility with high yields of the desired products using various types of primary alcohols, including aryl/hetero aryl methanols, and aliphatic alcohols as alkylating agents. The catalyst Ni NPs@N−C was synthesizedvia high‐temperature pyrolysis strategy, using ZIF‐8 as the sacrificial template. The Ni NPs@N−C catalyst was characterized by XPS, HR‐TEM, HAADF‐STEM, XRD and ICP‐MS. The catalyst is stable even in the air at room temperature, displayed excellent activity and could be recycled 5 times without appreciable loss of its activity. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
14. Therapy‐Related Acute Promyelocytic Leukemia: Case Series and Current Insights.
- Author
-
Bruzzese, Antonella, Martino, Enrica Antonia, Labanca, Caterina, Mendicino, Francesco, Lucia, Eugenio, Olivito, Virginia, Morelli, Rosellina, Rossi, Teresa, Neri, Antonino, Morabito, Fortunato, Gentile, Massimo, and Vigna, Ernesto
- Subjects
- *
ACUTE promyelocytic leukemia , *ACUTE myeloid leukemia , *ALKYLATING agents , *MALE breast cancer , *DNA topoisomerase II - Abstract
ABSTRACT Therapy‐related acute promyelocytic leukemia (t‐APL) is rare and often linked to previous treatment with alkylating agents or topoisomerase II inhibitors. This report describes three cases of t‐APL treated at the Haematology Department of Cosenza Hospital between 2022 and 2024, which occurred after alkylating agents and exemestane, alkylating agents and radiation therapy, alkylating agents, taxane, and checkpoint inhibitor, respectively. Each case was managed with a different therapeutic approach. The first case involved a 71‐year‐old man with colorectal and breast cancer, who developed low‐risk t‐APL and achieved complete remission (CR) with ATRA alone. A second 71‐year‐old man case with colorectal cancer developed high‐risk t‐APL with PML/RARA and FLT3‐ITD fusion transcripts; he achieved CR with idarubicin and ATRA despite severe sepsis and acute heart failure. The third case involved a 74‐year‐old man with lung squamous cell carcinoma who developed intermediate‐risk t‐APL following chemoimmunotherapy but unfortunately succumbed to pseudotumor cerebri complications during induction therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. A Modified Arbuzov‐Michalis Reaction for Selective Alkylation of Nucleophiles.
- Author
-
Wang, Jia‐Xi, Chen, Mu‐Qiu, Zhang, Yang, Han, Bo, Mou, Ze‐Dong, Feng, Xitong, Zhang, Xia, and Niu, Dawen
- Subjects
- *
ALKYLATING agents , *ETHYL acrylate , *ALKYLATION , *BIOCHEMICAL substrates , *NATURAL products - Abstract
The alkylation of nucleophiles is among the most fundamental and well‐developed transformations in chemistry. However, to achieve selective alkylation of complex substrates remains a nontrivial task. We report herein a general and selective alkylation method without using strong acids, bases, or metals. In this method, the readily available phosphinites/phosphites, in combination with ethyl acrylate, function as effective alkylating agents. Various nucleophilic groups, including alcohols, phenols, carboxylic acids, imides, and thiols can be alkylated. This method can be applied in the late‐stage alkylation of natural products and pharmaceutical agents, achieving chemo‐ and site‐selective modification of complex substrates. Experimental studies indicate the relative reactivity of a nucleophile depends on its acidity and its steric environment. Mechanistic studies suggest the reaction pathway resembles that of the Arbuzov‐Michalis reaction. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. MGMT methylation and its prognostic significance in inoperable IDH-wildtype glioblastoma: the MGMT-GBM study.
- Author
-
Ghimire, Prajwal, Kamaludin, Ahmad, Palau, Berta F., Lavrador, Jose P., Gullan, Richard, Vergani, Francesco, Bhangoo, Ranjeev, and Ashkan, Keyoumars
- Subjects
- *
O6-Methylguanine-DNA Methyltransferase , *ALKYLATING agents , *OVERALL survival , *SURVIVAL rate , *SURVIVAL analysis (Biometry) - Abstract
Introduction: The methylation of the O6-Methylguanine-DNA Methyltransferase (MGMT) promoter is a valid biomarker for predicting response to therapy with alkylating agents and, independently, prognosis in IDH-wildtype(IDH-w) glioblastoma. We aim to study the impact of its methylation in overall survival of the unresectable IDH-w glioblastoma undergoing biopsy and systemic treatment. Methods: We collected six-year retrospective (2017–2023) data at a quaternary neurosurgery center for patients undergoing biopsy as the only surgical procedure for an unresectable IDH wildtype glioblastoma. Data was collected from patient records including neuro-oncology multidisciplinary team meeting (MDT) documentation. Patients were grouped into categories according to different types of treatment received after biopsy (no treatment, chemotherapy (CT), radiotherapy (RT), chemoradiotherapy (CRT), chemoradiotherapy with adjuvant temozolomide (CRT with adjuvant TMZ), EORTC-NCIC protocol followed by second line treatment) and according to methylation status (unmethylated (< 5%), borderline methylated (5–15%) and strongly methylated (> 15%)). Survival analysis was performed. Results: 166 glioblastoma IDH wildtype patients were included in the study with mean age of 62.5 years (M: F = 1.5: 1). 70 (49.3%) patients had unmethylated MGMT status (< 5%), 29 (20.4%) patients had borderline methylated MGMT status (5–15%) and 43 (30.2%) patients had methylated MGMT status (> 15%). 36 (25.3%) patients did not receive any treatment post biopsy, 13 (9.1%) received CT only, 27 (19%) RT only, 12 (8.4%) CRT, 33 (23.2%) CRT with adjuvant TMZ, whereas 21 (14.7%) received EORTC-NCIC protocol along with second line treatment. In biopsy only group, there was no notable difference in survival outcomes among the different methylation statuses. For biopsy and any-other-form-of-treatment methylated groups showed a distinct trend of better survival compared to the borderline or unmethylated groups. Overall, methylated patients had better survival as compared to unmethylated or borderline groups. Conclusion: Methylated MGMT status are predictors for better overall survival in unresectable IDH wildtype glioblastoma patients undergoing biopsy and treatment regardless of the treatment modality. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. The Synergistic Combination of Curcumin and Polydatin Improves Temozolomide Efficacy on Glioblastoma Cells.
- Author
-
Serafino, Annalucia, Krasnowska, Ewa Krystyna, Romanò, Sabrina, De Gregorio, Alex, Colone, Marisa, Dupuis, Maria Luisa, Bonucci, Massimo, Ravagnan, Giampietro, Stringaro, Annarita, and Fuggetta, Maria Pia
- Subjects
- *
ANTINEOPLASTIC agents , *ALKYLATING agents , *CELL cycle , *TEMOZOLOMIDE , *OVERALL survival - Abstract
Glioblastoma (GBL) is one of the more malignant primary brain tumors; it is currently treated by a multimodality strategy including surgery, and radio- and chemotherapy, mainly consisting of temozolomide (TMZ)-based chemotherapy. Tumor relapse often occurs due to the establishment of TMZ resistance, with a patient median survival time of <2 years. The identification of natural molecules with strong anti-tumor activity led to the combination of these compounds with conventional chemotherapeutic agents, developing protocols for integrated anticancer therapies. Curcumin (CUR), resveratrol (RES), and its glucoside polydatin (PLD) are widely employed in the pharmaceutical and nutraceutical fields, and several studies have demonstrated that the combination of these natural products was more cytotoxic than the individual compounds alone against different cancers. Some of us recently demonstrated the synergistic efficacy of the sublingual administration of a new nutraceutical formulation of CUR+PLD in reducing tumor size and improving GBL patient survival. To provide some experimental evidence to reinforce these clinical results, we investigated if pretreatment with a combination of CUR+PLD can improve TMZ cytotoxicity on GBL cells by analyzing the effects on cell cycle, viability, morphology, expression of proteins related to cell proliferation, differentiation, apoptosis or autophagy, and the actin network. Cell viability was assessed using the MTT assay or a CytoSmart cell counter. CalcuSyn software was used to study the CUR+PLD synergism. The morphology was evaluated by optical and scanning electron microscopy, and protein expression was analyzed by Western blot. Flow cytometry was used for the cell cycle, autophagic flux, and apoptosis analyses. The results provide evidence that CUR and PLD, acting in synergy with each other, strongly improve the efficacy of alkylating anti-tumor agents such as TMZ on drug-resistant GBL cells through their ability to affect survival, differentiation, and tumor invasiveness. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. Gender-Specific Prognostic Impact of Treosulfan Levels in High-Dose Chemotherapy for Multiple Myeloma.
- Author
-
Heini, Alexander D., Kammermann, Karin, Bacher, Ulrike, Jeker, Barbara, Hayoz, Michael, Aebi, Yolanda, Largiadèr, Carlo R., Nilius, Henning, and Pabst, Thomas
- Subjects
- *
MULTIPLE myeloma , *HEMATOPOIETIC stem cell transplantation , *DRUG toxicity , *GENDER specific care , *SEX distribution , *TREATMENT effectiveness , *RETROSPECTIVE studies , *CANCER chemotherapy , *ETHERS , *PROGRESSION-free survival , *ALKYLATING agents , *OVERALL survival - Abstract
Simple Summary: High-dose chemotherapy (HDCT) with autologous stem cell transplantation (ASCT) is a standard treatment for multiple myeloma (MM). This study examines gender-specific differences in response to treosulfan and melphalan (TreoMel) HDCT. Data from 112 MM patients treated at a single center were analyzed for outcomes such as response rate, progression-free survival (PFS), overall survival (OS), and toxicities. Results revealed that females had significantly higher treosulfan exposure than males, as shown by peak levels and area under the curve (AUC). Notably, higher treosulfan exposure in females was associated with increased mortality, with those having an AUC > 900 mg*h/L showing shorter OS. However, treosulfan exposure did not affect PFS in females, and no significantly increased mortality risk was observed in males. These findings suggest that higher treosulfan doses increase toxicity in females without improving outcomes, supporting the need for further investigation into lower dosing for female MM patients. Introduction: The growing body of evidence around sexual and gender dimorphism in medicine, particularly in oncology, has highlighted differences in treatment response, outcomes, and side effects between males and females. Differences in drug metabolism, distribution, and elimination, influenced by factors like body composition and enzyme expression, contribute to these variations. Methods: We retrospectively analyzed data of 112 multiple myeloma (MM) patients treated with first-line high-dose chemotherapy (HDCT) with treosulfan and melphalan (TreoMel) followed by autologous stem cell transplantation (ASCT) at a single academic center between January 2020 and August 2022. We assessed response rate, progression-free survival (PFS), overall survival (OS), and toxicities in relation to gender and treosulfan exposure. Results: Our analysis revealed significant gender-specific differences in treosulfan exposure. Females had higher peak levels (343.8 vs. 309.0 mg/L, p = 0.0011) and area under the curve (AUC) (869.9 vs. 830.5 mg*h/L, p = 0.0427) compared to males. Higher treosulfan exposure was associated with increased mortality in females but not in males. Females with treosulfan AUC > 900 mg*h/L had significantly shorter overall survival, while PFS was unaffected by treosulfan exposure. Conclusion: Our study demonstrates that female patients undergoing TreoMel HDCT have higher treosulfan exposure than males and that females with higher levels are at increased risk for toxicity and adverse outcomes. These data suggest that higher treosulfan doses do not confer a benefit in terms of better outcomes for females. Therefore, exploring lower treosulfan doses for female MM patients undergoing TreoMel HDCT may be warranted to mitigate toxicity and improve outcomes. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
19. Retrospective study on pomalidomide‐PACE as a salvage regimen in aggressive relapsed and refractory multiple myeloma.
- Author
-
Gezer, Deniz, Nogueira, Melanie Schmitt, Kirschner, Martin, Brümmendorf, Tim H., Müller‐Tidow, Carsten, Goldschmidt, Hartmut, Raab, Marc S., and Giesen, Nicola
- Subjects
- *
PLASMA cell leukemia , *ALKYLATING agents , *CHIMERIC antigen receptors , *EXTRAMEDULLARY diseases , *MULTIPLE myeloma - Abstract
Objectives: Despite major advances in treatment options for multiple myeloma (MM), patients refractory to the main drug classes and those with aggressive, especially extramedullary disease, still face a dismal outcome. For these patients, effective therapeutic options are urgently warranted. Methods: In this retrospective study, we report on the safety and efficacy of the intensive combination regimen of pomalidomide plus cisplatin, doxorubicin, cyclophosphamide, and etoposide (Pom‐PACE) in patients with relapsed refractory MM (RRMM) or plasma cell leukemia (PCL). A study population of 20 consecutive patients treated with Pom‐PACE at two academic centers was included for analysis. All patients had to have a confirmed relapse according to International Myeloma Working Group criteria and adequate organ function prior to the start of therapy. Data were collected by reviewing medical charts. Exploratory analyses were performed with regard to efficacy and safety. Results: Patients were heavily pretreated with a median number of four prior therapies (range: 1–10). All patients were exposed to immunomodulators, proteasome inhibitors, and alkylating agents, 80% were double‐class refractory, 40% were triple‐class refractory. Extramedullary MM or PCL were present in 15 patients (75%). Overall response rate (ORR) was 68%, with 31% achieving at least a very good partial response. Responses were achieved rapidly with an ORR of 64% after one cycle. Median progression‐free survival was 8.9 months (0.92–not reached [NR]) and median overall survival was 11.8 months (3–40.6). Pom‐PACE was associated with significant toxicity. All evaluable patients experienced Grade 4 hematological toxicity. However, no treatment related mortality was observed. Conclusion: Pomalidomide‐PACE was able to induce rapid responses in heavily pretreated, aggressive RRMM with a manageable toxicity profile and therefore offers an effective salvage regimen and a potential bridging strategy to further treatment options such as chimeric antigen receptor T‐cell therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
20. Non-Infectious Uveitis and Pregnancy, is There an Optimal Treatment? Uveitis Course and Safety of Uveitis Treatment in Pregnancy.
- Author
-
Ting, Magdalene Yin Lin, Vega-Tapia, Fabian, Anguita, Rodrigo, Cuitino, Loreto, Valenzuela, Rodrigo A., Salgado, Felipe, Valenzuela, Omar, Ibañez, Sebastian, Marchant, Ruben, and Urzua, Cristhian A.
- Subjects
- *
CERTOLIZUMAB pegol , *EYE inflammation , *PREGNANCY outcomes , *MATERNALLY acquired immunity , *ALKYLATING agents - Abstract
In pregnancy, a plethora of factors causes changes in maternal immunity. Uveitis flare-ups are more frequent in the first trimester and in undertreated patients. Management of non-infectious uveitis during pregnancy remains understudied. A bibliographic review to consolidate existing evidence was performed by a multidisciplinary group of Ophthalmologists, Gynaecologists and Rheumatologists. Our group recommends initial management with minimum-required doses of corticosteroids, preferably locally, to treat intraocular inflammation whilst ensuring good neonatal outcomes. If ineffective, clinicians should consider addition of Cyclosporine, Azathioprine or Certolizumab pegol, which are seemingly safe in pregnancy. Other therapies (such as Methotrexate, Mycophenolate Mofetil and alkylating agents) are teratogenic or have a detrimental effect on the foetus. Furthermore, careful multidisciplinary preconception discussions and close follow-up are recommended, monitoring for flare-ups and actively tapering medication doses, with a primary endpoint focused on protecting ocular tissues from inflammation, whilst giving minimal risk of poor pregnancy and foetal outcomes. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
21. Rh(III)-catalyzed aldehydic and aryl C–H alkylation with cyclopropanols via C–H/C–C bond activation.
- Author
-
Dash, Om Prakash, Garg, Divya, and Volla, Chandra M. R.
- Subjects
- *
ALKYLATING agents , *ANILINE derivatives , *CYCLOPROPANOL , *NATURAL products , *ALKYLATION - Abstract
Herein, we report Rh(III) catalyzed aldehydic or aryl C–H alkylation via C–C bond activation of cyclopropanols, facilitating the synthesis of β-functionalized ketones. The protocol employs cyclopropanol as the alkylating agent with 2-aminobenzaldehyde or aniline derivatives to access a variety of unsymmetrical 1,4-diketones or β-aryl ketones, respectively. The practicality of these transformations is showcased through the modification of natural products, gram-scale synthesis, broad substrate scope and postfunctionalizations. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
22. Symmetric Aromatic Amidoalkylations of Triphenylenes.
- Author
-
Lorenzetto, Tommaso, Berton, Giacomo, Castellini, Francesco, Casson, Gabriele, Scarso, Alessandro, and Fabris, Fabrizio
- Subjects
- *
ALKYLATING agents , *CARBOXAMIDES , *ALKYLATION , *HYDROXYMETHYL compounds - Abstract
Triphenylene derivatives are highly investigated for their electronic, supramolecular and photophysical properties, but the direct modification of the central aromatic core is particularly challenging especially in the internal positions 1, 4, 5, 8, 9, and 12. Herein we present an efficient alkylation method of 2,3,6,7,10,11‐hexasubstituted triphenylene derivatives leading to tris‐alkylated C3‐symmetric derivatives in good yields using N‐(hydroxymethyl)carboxamide or N‐(alkoxylmethyl)carboxamide alkylating agents. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
23. The tumor microenvironment in therapy resistance.
- Author
-
Garcia, Guadalupe G., Schmidt, Christopher J., and Hajal, Cynthia
- Subjects
IMMUNE checkpoint inhibitors ,ALKYLATING agents ,SARCOMA ,TUMOR microenvironment ,CANCER relapse - Abstract
Chemotherapy alone or in conjunction with surgery and radiation is often used to treat various cancer types. While effective at treating some tumors, the response varies across patients with different malignancies. For some cancers, such as glioblastoma, ovarian cancer, and soft tissue sarcoma, 85%-100% of patients experience cancer recurrence and develop chemotherapy resistance, which often leads to worse prognoses. These alarming statistics highlight an urgent need to better understand the landscape of therapy resistance in cancer, in order to develop improved treatment strategies and prevent recurrence. A central focus has been the investigation of resistant tumor subclones and whether the use of different alkylating agents and/or immune checkpoint inhibitors can ablate different clones. However, very little effort has been directed towards studies of the tumor microenvironment, a complex ecosystem of blood vessels, fibroblasts, immune cells, signaling molecules, and extracellular matrix, in the context of therapy resistance. In this perspective, we provide an overview of different platforms, tools, and techniques that have been developed and used to identify tumor microenvironment alterations due to therapy resistance. We also address potential therapeutic strategies that involve components of the tumor milieu and have been identified and tested to overcome treatment-induced resistance. Identifying microenvironmental changes post-resistance presents opportunities for new targeted treatment strategies. The current state of the literature suggests a dire need for more engineered models that probe specific microenvironment contributors to therapy resistance or ways in which the tumor tissue can be harnessed to mitigate resistance. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
24. Dual-Action Therapeutics: DNA Alkylation and Antimicrobial Peptides for Cancer Therapy.
- Author
-
Andrés, Celia María Curieses, Pérez de la Lastra, José Manuel, Munguira, Elena Bustamante, Andrés Juan, Celia, and Pérez-Lebeña, Eduardo
- Subjects
- *
THERAPEUTIC use of antineoplastic agents , *CELL proliferation , *IMMUNOTHERAPY , *MECHLORETHAMINE , *ANTIMICROBIAL peptides , *DNA , *IMMUNODIAGNOSIS , *CANCER chemotherapy , *MONOCLONAL antibodies , *NITROGEN mustards , *CELL lines , *DNA damage , *CELL death , *MOLECULAR structure , *CYTOCHROME P-450 , *TUMORS , *GENETIC mutation , *CELL surface antigens - Abstract
Simple Summary: Conventional cancer treatments, based on chemotherapy and radiotherapy, are often effective but suffer from serious side effects and a potential risk of resistance. Dual therapies, combining DNA alkylating agents and antimicrobial peptides, are generating great interest. Within chemotherapies, a frequently used mechanism is DNA alkylation, inducing DNA damage and subsequent cell death. Antimicrobial peptides, in turn, have demonstrated their efficacy as anticancer agents due to their ability to selectively alter cancer cell membranes. In this review, our aim has been to explore the synergistic potential of these two therapeutic modalities when used together. Cancer remains one of the most difficult diseases to treat, requiring continuous research into innovative therapeutic strategies. Conventional treatments such as chemotherapy and radiotherapy are effective to a certain extent but often have significant side effects and carry the risk of resistance. In recent years, the concept of dual-acting therapeutics has attracted considerable attention, particularly the combination of DNA alkylating agents and antimicrobial peptides. DNA alkylation, a well-known mechanism in cancer therapy, involves the attachment of alkyl groups to DNA, leading to DNA damage and subsequent cell death. Antimicrobial peptides, on the other hand, have been shown to be effective anticancer agents due to their ability to selectively disrupt cancer cell membranes and modulate immune responses. This review aims to explore the synergistic potential of these two therapeutic modalities. It examines their mechanisms of action, current research findings, and the promise they offer to improve the efficacy and specificity of cancer treatments. By combining the cytotoxic power of DNA alkylation with the unique properties of antimicrobial peptides, dual-action therapeutics may offer a new and more effective approach to fighting cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
25. Friedel–crafts alkylations of indoles, furans, and thiophenes with arylmethyl acetates promoted by trimethylsilyl trifluoromethanesulfonate.
- Author
-
Xia, Helen L., Zhou, Eric, Bicalho, Bianca, and Downey, C. Wade
- Subjects
- *
ALKYLATING agents , *THIOPHENES , *BENZOFURANS , *ALKYLATION , *INDOLE compounds - Abstract
Various indoles undergo Friedel–Crafts alkylation at the 3-position when treated with secondary arylmethyl acetates in the presence of trimethylsilyl trifluoromethanesulfonate (TMSOTf) and 2,6-lutidine. Tertiary benzylic alcohols are also effective alkylating agents, undergoing TMSOTf-promoted activation to yield alkylated indoles that feature quaternary centers. A related study shows that benzofuran reacts under similar conditions to yield 2-alkylated benzofurans when treated with similar alkylating agents, and the reaction has been extended to include other representative heteroarenes. Yields for these Friedel–Crafts alkylation products range from 43%–99% over a wide range of substrate combinations. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
26. Comprehensive understanding of the adverse effects associated with temozolomide: a disproportionate analysis based on the FAERS database.
- Author
-
Yusen Zhou, Peng Jia, Yuting Fang, Wei Zhu, Yong Gong, Tianyu Fan, and Jiangliu Yin
- Subjects
DRUG side effects ,ALKYLATING agents ,TEMOZOLOMIDE ,APLASTIC anemia ,WATER-electrolyte imbalances - Abstract
Background: Temozolomide, which is the standard drug for glioma treatment, has several Adverse events (AEs) in the treatment of gliomas and other tumors that are not yet fully understood. This is due to the pharmacological nature of the alkylating agent. A significant proportion of these effects have not been systematically documented or reported. Methods: We selected data from the United States FDA Adverse Event Reporting System (FAERS) database from the first quarter of 2004 to the fourth quarter of 2023. Four algorithms were used for disproportionate analysis, with the objective of assessing the association between temozolomide and related adverse events. Results: In this study, 20,079,906 case reports were collected from the FAERS database, of which 15,152 adverse events related to temozolomide were reported. A total of 352 preferred terms (PTs) and 24 system organ classes (SOCs) that were significantly disproportionally related to the four algorithms were included. The SOCs included blood and lymphatic system disorders (χ² = 18,220.09, n = 4,325); skin and subcutaneous tissue disorders (χ² = 408.06, n = 1,347); investigations (χ² = 639.44, n = 3,925); musculoskeletal and connective tissue disorders (χ² = 1,317.29, n = 588); and psychiatric disorders (χ² = 1,098.47, n = 877). PT levels were screened for adverse drug reaction signals consistent with drug inserts, such as anemia, thrombocytopenia, liver function abnormalities, nausea and vomiting, as well as rarely reported adverse drug reactions, such as aplastic anemia, myelodysplastic syndromes, electrolyte disorders, cerebral edema, and high-frequency mutations. Conclusion: The results of our investigation demonstrated both adverse effects that had been reported and a multitude of unreported adverse effects that were serious in nature and lacked a clear cause. These novel findings suggest that more attention should be given to the clinical conditions of patients after treatment to provide a more comprehensive perspective and understanding for further clarifying the safety of temozolomide. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
27. Identification of a novel DNA oxidative damage repair pathway, requiring the ubiquitination of the histone variant macroH2A1.1.
- Author
-
Ouararhni, Khalid, Mietton, Flore, Sabir, Jamal S. M., Ibrahim, Abdulkhaleg, Molla, Annie, Albheyri, Raed S., Zari, Ali T., Bahieldin, Ahmed, Menoni, Hervé, Bronner, Christian, Dimitrov, Stefan, and Hamiche, Ali
- Subjects
- *
POLY(ADP-ribose) polymerase , *DNA damage , *ALKYLATING agents , *GENETIC transcription regulation , *CELL survival , *DNA repair , *CHROMATIN - Abstract
Background: The histone variant macroH2A (mH2A), the most deviant variant, is about threefold larger than the conventional histone H2A and consists of a histone H2A-like domain fused to a large Non-Histone Region responsible for recruiting PARP-1 to chromatin. The available data suggest that the histone variant mH2A participates in the regulation of transcription, maintenance of heterochromatin, NAD+ metabolism, and double-strand DNA repair. Results: Here, we describe a novel function of mH2A, namely its implication in DNA oxidative damage repair through PARP-1. The depletion of mH2A affected both repair and cell survival after the induction of oxidative lesions in DNA. PARP-1 formed a specific complex with mH2A nucleosomes in vivo. The mH2A nucleosome-associated PARP-1 is inactive. Upon oxidative damage, mH2A is ubiquitinated, PARP-1 is released from the mH2A nucleosomal complex, and is activated. The in vivo-induced ubiquitination of mH2A, in the absence of any oxidative damage, was sufficient for the release of PARP-1. However, no release of PARP-1 was observed upon treatment of the cells with either the DNA alkylating agent MMS or doxorubicin. Conclusions: Our data identify a novel pathway for the repair of DNA oxidative lesions, requiring the ubiquitination of mH2A for the release of PARP-1 from chromatin and its activation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
28. Teratogenicity is more likely a function of primary and secondary pharmacology than caused by chemically reactive metabolites: a critical evaluation of 40 years of scientific research.
- Author
-
Smith, Dennis A.
- Subjects
- *
ALKYLATING agents , *ANIMAL experimentation , *ACID derivatives , *RETINOIDS , *THALIDOMIDE - Abstract
The number of therapeutic drugs known to be human teratogens is actually relatively small. This may reflect the rigorous animal testing and well defined labelling. Some of these drugs were identified to have reactive metabolites and this has been postulated, historically, to be their teratogenic mechanism. These drugs include thalidomide, various anticonvulsants and retinoic acid derivatives. Many of these experiments were conducted in a period where chemically reactive metabolites were being intensely investigated and associated with all forms of toxicity. The legacy of this is that these examples are routinely cited as well established mechanisms. Examination of mechanism leads to the conclusion that the teratogenicity in humans of these compounds is likely due to the primary and secondary pharmacology of the parent drug and stable circulating metabolites and that association of reactive metabolites to this toxicity is unwarranted. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
29. Protective effects of Withania somnifera against cyclophosphamide-induced testicular damage in rats.
- Author
-
Jafari, Mehrana, Akbari, Ahmad, Esmailpour, Zeynab, Nadi, Zahra, and Baazm, Maryam
- Subjects
- *
GENITALIA , *ALKYLATING agents , *SEMINIFEROUS tubules , *GENE expression , *WITHANIA somnifera - Abstract
Objective: Cyclophosphamide (CP) is an alkylating agent commonly used in cancer treatment. It is known to have detrimental effects on the reproductive system, including the potential to cause infertility. Recently, herbal remedies have gained traction as a complementary approach to addressing these side effects. In this study, our goal was to investigate whether the aqueous-alcoholic extract of Withania somnifera (WS) could mitigate the adverse impacts of CP on testicular tissue. Methods: Animals were randomly assigned to one of the following groups: control, WS (500 mg/kg), CP (100 mg/kg), CP+WS pre-treatment, and CP+WS post-treatment. WS was administered orally through gavage for 1 month. We assessed sperm parameters, testicular histopathology, and the expression of the Bax and Bcl2 genes in the experimental groups. Results: Sperm parameters (including count, viability, and motility), the number of spermatogonia, the seminiferous tubule diameter, and Bcl2 gene expression, significantly decreased after CP injection (p<0.05). Conversely, the number of immotile sperm and Bax gene expression significantly increased (p<0.05). Treatment with WS, especially when administered as a pre-treatment, ameliorated the sperm parameters, histological alterations, and the expression of apoptosis-related genes (p<0.05). Conclusion: The data suggest that WS may mitigate the detrimental effects of CP on testicular tissue by reducing apoptosis. Consequently, WS has the potential to be used as an adjunctive therapy to reduce the complications associated with CP treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
30. A Comparative Analysis of Chemotherapeutic Administrations in Inhibiting Glioblastoma Multiforme Cellular Growth Utilizing an Integration of Differentiation-Based Growth Models and Pharmacokinetic Equations.
- Author
-
Gowda, Anagha Chethan
- Subjects
- *
ALKYLATING agents , *BRAIN tumors , *GLIOBLASTOMA multiforme , *TUMOR growth , *CELL growth - Abstract
Glioblastoma multiforme, a disease of grade IV astrocytoma, accounts for 60-70% of malignant brain tumors.? Despite its low survival rate, little is known about GBM and its treatment. The purpose of this study was to assess the comparative effectiveness of the most common drugs (temozolomide, carmustine, bevacizumab) in reducing tumor growth within GBM chemotherapy. It was hypothesized that due to their properties as alkylating agents inhibiting growth genes within GBM, temozolomide and carmustine would provide the most reduction in cellular growth mathematically.² VisualPDE simulations were used to model administration of temozolomide, carmustine, bevacizumab, and none in chemotherapy and provide the cellular GBM counts at any given time within five months (60 time points were used). Results showed cell growth reduction by temozolomide and carmustine, but most drastically by bevacizumab, and upon further analysis, a One-Way ANOVA yielded that the difference between at least two chemotherapeutic administrations were statistically significant (F(3, 236)=281.049, p<0.001). A post-hoc Tukey HSD test revealed a significant difference between no administration and each chemotherapeutic administration (TMZ: p<0.001, BCNU: p=0.046, BEV: p<0.001), as well as significant differences compared to bevacizumab, which was drastically more effective than TMZ and BCNU (p<0.001, p<0.001). Thus, it was concluded that BEV provided the optimal treatment in slowing GBM growth with the mathematical model through its ability to regulate proteins and drug resistance, allowing for its integration in future chemotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
31. Study of Liposomes Containing Extract from the Leaves of Protium heptaphyllum (Aubl.) March in Animals Submitted to a Mutagenic Model Induced by Cyclophosphamide.
- Author
-
Patias, Naiéle Sartori, Sinhorin, Valéria Dornelles Gindri, Ferneda, Ana Júlia Lopes Braga, Ferneda, João Maurício Andrade, Sugui, Marina Mariko, Ferrarini, Stela Regina, Bomfim, Gisele Facholi, Lopes, Joaz Wellington, Antoniassi, Nadia Aline Bobbi, Cavalheiro, Larissa, Domingues, Nelson Luís de Campos, and Sinhorin, Adilson Paulo
- Subjects
- *
ALKYLATING agents , *LABORATORY mice , *SUPEROXIDE dismutase , *ALKALINE phosphatase , *PLANT extracts , *LIPOSOMES - Abstract
Simple Summary: In this study, the protective effect of Protium heptaphyllum (P. heptaphyllum), a plant with a high flavonoid content, was analyzed against damage caused by cyclophosphamide (CPA), a chemotherapy drug known for its adverse effects. Using liposomes to transport the plant extract, an experiment was carried out with male Swiss mice that received the extract for 14 days before CPA. The results demonstrated that P. heptaphyllum liposomes reduced DNA damage and oxidative stress, evidenced by the increase in antioxidant enzymes such as superoxide dismutase (SOD) and glutathione peroxidase (GPx). There were also improvements in liver and kidney indicators, suggesting organ protection. In summary, these results highlight the potential of P. heptaphyllum liposomes as a natural chemical protection agent that can help reduce the side effects of cancer treatment, while providing antioxidant benefits. Cyclophosphamide (CPA) is an alkylating agent used as a chemotherapy agent in the treatment of cancer, but it has immunosuppressive effects. Protium heptaphyllum (P. heptaphyllum) is a plant rich in triterpenes and flavonoids, with some bioactive and therapeutic properties presented in the literature. Thus, the present study aimed to investigate the chemoprotective potential of P. heptaphyllum extract inserted into liposomes against oxidative damage chemically induced by CPA. Male Swiss mice received 1.5 mg/kg of P. heptaphyllum liposomes as a pre-treatment for 14 consecutive days (via gavage) and 100 mg/kg of CPA in a single dose (via intraperitoneal) on the 15th day. After the experimental period, blood and organ samples were collected for histopathological and biochemical analyses, including superoxide dismutase (SOD), glutathione peroxidase (GPx), catalase (CAT), glutathione S-transferase (GST), reduced glutathione (GSH), thiobarbituric acid reactive substances (TBARS), ascorbic acid (ASA), carbonyl protein, cytokine measurement, and micronucleus testing. The results showed that liposomes containing P. heptaphyllum extract have an antimutagenic effect against damage induced to DNA by CPA, and that they also protect against oxidative stress, as verified by the increase in the antioxidant enzymes SOD and GPx. The improvement in alkaline phosphatase and creatinine markers suggests a beneficial effect on the liver and kidneys, respectively. However, the depletion of GSH in the liver and brain suggests the use of antioxidants for the metabolism of molecules generated in these tissues. In general, these data show good prospects for the use of P. heptaphyllum liposomes as a cancer chemoprotective agent, as well as possible antioxidant action, conceivably attributed to the flavonoids present in the plant extract. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
32. Interindividual variation in ovarian reserve after gonadotoxic treatment in female childhood cancer survivors – a genome-wide association study: results from PanCareLIFE.
- Author
-
van der Perk, M.E. Madeleine, Broer, Linda, Yasui, Yutaka, Laven, Joop S.E., Robison, Leslie L., Tissing, Wim J.E., Versluys, Birgitta, Bresters, Dorine, Kaspers, Gertjan J.L., Lambalk, Cornelis B., Overbeek, Annelies, Loonen, Jacqueline J., Beerendonk, Catharina C.M., Byrne, Julianne, Berger, Claire, Clemens, Eva, van Dulmen-den Broeder, Eline, Dirksen, Uta, van der Pal, Helena J., and de Vries, Andrica C.H.
- Subjects
- *
GENOME-wide association studies , *SINGLE nucleotide polymorphisms , *TOTAL body irradiation , *ALKYLATING agents , *GENETIC variation , *OVARIAN cancer , *OVARIAN reserve ,GONADAL diseases - Abstract
To discover new variants associated with low ovarian reserve after gonadotoxic treatment among adult female childhood cancer survivors using a genome-wide association study approach. Genome-wide association study. Not applicable. A discovery cohort of adult female childhood cancer survivors from the pan-European PanCareLIFE cohort (n = 743; median age: 25.8 years), excluding those who received bilateral ovarian irradiation, bilateral oophorectomy, central nervous system or total body irradiation, or stem cell transplantation. Replication was attempted in the US-based St. Jude Lifetime Cohort (n = 391; median age: 31.3 years). Female childhood cancer survivors are at risk of therapy-related gonadal impairment. Alkylating agents are well-established risk factors, and the interindividual variability in gonadotoxicity may be explained by genetic polymorphisms. Data were collected in real-life conditions, and cyclophosphamide equivalent doses were used to quantify alkylation agent exposure. Anti-Müllerian hormone (AMH) levels served as a proxy for ovarian function, and the findings were combined in a meta-analysis. Three genome-wide significant (<5.0 × 10−8) and 16 genome-wide suggestive (<5.0 × 10−6) loci were associated with log-transformed AMH levels, adjusted for cyclophosphamide equivalent dose of alkylating agents, age at diagnosis, and age at study in the PanCareLIFE cohort. On the basis of the effect allele frequency (EAF) (>0.01 if not genome-wide significant), and biologic relevance, 15 single nucleotide polymorphisms were selected for replication. None of the single nucleotide polymorphisms were statistically significantly associated with AMH levels. A meta-analysis indicated that rs78861946 was associated with borderline genome-wide statistical significance (reference/effect allele: C/T; effect allele frequency: 0.04, beta (SE): −0.484 (0.091). This study found no genetic variants associated with a lower ovarian reserve after gonadotoxic treatment because the findings of this genome-wide association study were not statistically significant replicated in the replication cohort. Suggestive evidence for the potential importance of 1 variant is briefly discussed, but the lack of statistical significance calls for larger cohort sizes. Because the population of childhood cancer survivors is increasing, large-scale and systematic research is needed to identify genetic variants that could aid predictive risk models of gonadotoxicity as well as fertility preservation options for childhood cancer survivors. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
33. CARDIAC SURVEILLANCE IN ONCOLOGY: A REVIEW OF CIRCULATING BIOMARKERS AND DIAGNOSIS METHODS IN CHEMOTHERAPY-INDUCED CARDIOTOXICITY.
- Author
-
ANCA, EMILIA, BANCIU, MANUELA, ROȘIORU, CORINA-LUMINIȚA, and DOBRE, CAMELIA
- Subjects
NATRIURETIC peptides ,HEART diseases ,ALKYLATING agents ,CARDIOVASCULAR system ,CARDIOTOXICITY ,HEART failure - Abstract
Copyright of Farmacia is the property of Societatea de Stiinte Farmaceutice Romania and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2024
- Full Text
- View/download PDF
34. Development and Application of a Slot-Blot Assay Using the Damage Sensing Protein Atl1 to Detect and Quantify O 6 -Alkylated Guanine Bases in DNA.
- Author
-
Yaakub, Hanum, Howell, Anthony, Margison, Geoffrey P., and Povey, Andrew C.
- Subjects
DNA alkylation ,HUMAN DNA ,ALKYLATING agents ,SCHIZOSACCHAROMYCES pombe ,TEMOZOLOMIDE ,METHYLGUANINE - Abstract
Humans are unavoidably exposed to numerous different mutagenic DNA alkylating agents (AAs), but their role in the initiation of cancers is uncertain, in part due to difficulties in assessing human exposure. To address this, we have developed a screening method that measures promutagenic O
6 -alkylguanines (O6 -AlkGs) in DNA and applied it to human DNA samples. The method exploits the ability of the Schizosaccharomyces pombe alkyltransferase-like protein (Atl1) to recognise and bind to a wide range of O6 -AlkGs in DNA. We established an Atl1-based slot-blot (ASB) assay and validated it using calf thymus DNA alkylated in vitro with a range of alkylating agents and both calf thymus and human placental DNA methylated in vitro with temozolomide (TMZ). ASB signals were directly proportional to the levels of O6 -meG in these controls. Pre-treatment of DNA with the DNA repair protein O6 -methylguanine–DNA methyltransferase (MGMT) reduced binding of Atl1, confirming its specificity. In addition, MCF 10A cells were treated with 500 μM TMZ and the extracted DNA, analysed using the ASB, was found to contain 1.34 fmoles O6 -meG/μg DNA. Of six human breast tumour DNA samples assessed, five had detectable O6 -AlkG levels (mean ± SD 1.24 ± 0.25 O6 -meG equivalents/μg DNA. This study shows the potential usefulness of the ASB assay to detect and quantify total O6 -AlkGs in human DNA samples. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
35. The Assessment of Methyl Methanesulfonate Absorption by Amphipods from the Environment Using Lux-Biosensors.
- Author
-
Novoyatlova, Uliana S., Kudryavtseva, Anna A., Bazhenov, Sergey V., Utkina, Anna A., Fomin, Vadim V., Nevmyanov, Shamil A., Zhoshibekova, Bagila S., Fedyaeva, Maria A., Kolobov, Mikhail Y., and Manukhov, Ilya V.
- Subjects
METHYL methanesulfonate ,ALKYLATING agents ,PHOTORHABDUS luminescens ,FISHERIES ,AMPHIPODA - Abstract
The ability of aquatic mesofauna representatives involved in trophic chains to sorb and accumulate toxicants is important for understanding the functioning of aquatic ecosystems and for fishing industry. This study investigated the capacity of marine amphipod Gammarus oceanicus and freshwater amphipods Eulimnogammarus vittatus and Gammarus lacustris to absorb the DNA-alkylating agent methyl methanesulfonate (MMS). The presence of alkylating agents in the environment and in the tissues of the amphipods was determined using whole-cell lux-biosensor Escherichia coli MG1655 pAlkA-lux, in which the luxCDABE genes from Photorhabdus luminescens, enabling the luminescence of the cell culture, are controlled by the P
alkA promoter of DNA glycosylase. It was shown that within one day of incubation in water containing MMS at a concentration above 10 μM, the amphipods absorbed the toxicant and their tissues produce more alkylation damage to biosensor cells than the surrounding water. Concentrations of MMS above 1 mM in the environment caused the death of the amphipods before the toxicant could be significantly concentrated in their tissues. The sensitivity and the capacity to absorb MMS were found to be approximately the same for the marine amphipod G. oceanicus and the freshwater amphipods E. vittatus and G. lacustris. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
36. Reduced Intensity Allogeneic HCT in Advanced Hematologic Malignancies w/T-Cell Depleted Graft
- Author
-
Orca Biosystems, Inc.
- Published
- 2024
37. Combination Therapy with Immune Checkpoint Inhibitors and Histone Deacetylase Inhibitors or Alkylating Agents
- Author
-
Joerger M, Koster KL, Janik T, and de Jong FA
- Subjects
histone deacetylase inhibitor ,checkpoint inhibitor ,alkylating agents ,synergy ,hematological malignancies ,solid tumors ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Markus Joerger,1 Kira-Lee Koster,1 Tomas Janik,2 Floris A de Jong3,4 1Department of Medical Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland; 2Research & Development Department, Mundipharma Research Limited, Cambridge, UK; 3Global Medical Affairs Department, Mundipharma Research Limited, Cambridge, UK; 4Medical Affairs Department, Exact Sciences International GmbH, Baar, SwitzerlandCorrespondence: Tomas Janik, Mundipharma Research Limited, Cambridge Science Park, Milton Road, Cambridge, UK, Email tomas.janik@mundipharma.czPurpose: Immune checkpoint inhibitors (CPIs) have been widely adopted in a number of early and advanced malignancies. Histone deacetylase inhibitors (HDACis) and alkylating agents (AAs) have been suggested to potentiate the actions of CPIs on tumor cells. We conducted a comprehensive literature review to explore the potential synergistic activity between CPIs, AAs, and HDACis.Patients and Methods: Clinical and non-clinical studies describing outcomes in patients with cancer receiving CPIs and either concomitant or sequential (pre- or post-CPI) AAs or HDACis were identified in PubMed using pre-defined search strings. Manual searches of key oncology congresses were similarly performed. All relevant articles and abstracts were manually screened for relevance, classified according to the specific anticancer agents used (CPIs, AAs, or HDACis), tumor entity, and whether treatment was concomitant or sequential.Results: Overall, 227 unique clinical studies across a range of tumor types, both solid tumors and hematological malignancies, were identified. One hundred and fifty-nine publications on Phase I and II clinical studies together with 41 publications on Phase III studies were examined. The most commonly investigated tumor types were melanoma, triple-negative breast cancer, non-small cell lung cancer, and Hodgkin lymphoma. The randomized clinical studies identified, all of which reported on the combination of a CPI with an AA, demonstrated superior outcomes in the combination arm compared with CPI or AA monotherapy. Similarly, combination therapy with CPIs and HDACis demonstrated promising activity.Conclusion: Sequential or concomitant administration of a CPI with an AA or an HDACi may improve outcomes for patients with a range of tumor types. There is a rationale to support further investigation into the potential for synergy between CPIs, alkylating agents and/or HDACis in both the non-clinical and clinical settings.Plain Language Summary: People being treated for cancer will often receive more than one drug at a time, and the concept of combining cancer drugs is frequently investigated as a potential opportunity to improve outcomes for patients. We reviewed the published literature for clinical trials and work undertaken in laboratories to explore whether combining targeted agents that stop cancer cells from multiplying (known as checkpoint inhibitors) with traditional chemotherapy that kills cancer cells could be a useful approach. We looked at evidence in publications where checkpoint inhibitors were used at the same time as chemotherapy, or given immediately before or after chemotherapy. The most important evidence came from clinical trials where outcomes for patients receiving combinations of treatment were directly compared with those from patients receiving a single treatment. These studies showed superior outcomes for patients who were treated with a combination of cancer drugs compared with patients receiving monotherapy. We also found evidence that adding another class of cancer drug, called histone deacetylase inhibitors, might sensitize tumors to checkpoint inhibitors. These findings provide a rationale for examining alkylating agents and/or histone deacetylase inhibitors combined with checkpoint inhibitors.Keywords: histone deacetylase inhibitor, checkpoint inhibitor, alkylating agents, synergy, hematological malignancies, solid tumors
- Published
- 2024
38. Green Synthesis of Diphenyl‐Substituted Alcohols Via Radical Coupling of Aromatic Alcohols Under Transition‐Metal‐Free Conditions.
- Author
-
Le, Ha V., Nguyen, Vy T. B., Le, Huy X., Nguyen, Tung T., Nguyen, Khoa D., Ho, Phuoc H., and Nguyen, Thuong T. H.
- Subjects
- *
BENZYL alcohol , *ALKYLATING agents , *RADICAL anions , *RADICALS (Chemistry) , *CHARGE exchange - Abstract
Alcohols are common alkylating agents and starting materials alternative to harmful alkyl halides. In this study, a simple, benign and efficient pathway was developed to synthesize 1,3‐diphenylpropan‐1‐ols
via theβ ‐alkylation of 1‐phenylethanol with benzyl alcohols. Unlike conventional borrowing hydrogen processes in which alcohols were activated by transition‐metal catalyzed dehydrogenation, in this work,t‐ BuONa was suggested to be a dual‐role reagent, namely, both base and radical initiator, for the radical coupling of aromatic alcohols. The cross‐coupling reaction readily proceeded under transition metal‐free conditions and an inert atmosphere, affording 1,3‐diphenylpropan‐1‐ol with an excellent yield. A good functional group tolerance in benzyl alcohols was observed, leading to the production of various phenyl‐substituted propan‐1‐ol derivatives in moderate‐to‐good yields. The mechanistic studies proposed that the reaction could involve the formation of reactive radical anions by base‐mediated deprotonation and single electron transfer. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
39. Molecular Factors Predicting Ovarian Chemotoxicity in Fertile Women: A Systematic Review.
- Author
-
Raimondo, Diego, Raffone, Antonio, Neola, Daniele, Genovese, Federica, Travaglino, Antonio, Aguzzi, Alberto, De Gobbi, Valeria, Virgilio, Agnese, Di Santo, Sara, Vicenti, Rossella, Magnani, Valentina, Guida, Maurizio, Pippucci, Tommaso, and Seracchioli, Renato
- Subjects
- *
GENETICS of disease susceptibility , *MEDICAL information storage & retrieval systems , *SEX hormones , *WOMEN , *REPRODUCTIVE health , *INFERTILITY , *OVARIAN tumors , *CANCER patients , *DESCRIPTIVE statistics , *CANCER chemotherapy , *SYSTEMATIC reviews , *MEDLINE , *ANTHRACYCLINES , *CANCER patient psychology , *OVARIAN reserve , *FERTILITY preservation , *ONLINE information services , *INDIVIDUALIZED medicine , *OVARIAN diseases , *BIOMARKERS , *OVARIES , *ALKYLATING agents , *CYCLOPHOSPHAMIDE - Abstract
Simple Summary: This article explores the impact of chemotherapy on ovarian function in female cancer survivors, emphasizing the importance of preserving fertility alongside cancer treatment. Chemotherapy-induced ovarian failure (CIOF) poses a significant concern, affecting patients' quality of life. The study aims to identify genetic markers predictive of CIOF through a systematic review of existing literature. The review identifies four relevant studies focusing on genetic factors associated with CIOF. Findings suggest potential associations between genetic variations, like CYP3A4*1B and GSTA1, and CIOF risk. Moreover, BRCA mutations may influence ovarian reserve recovery post-chemotherapy. While current assessment methods rely on biochemical tests and ultrasound imaging, genetic testing holds promise for personalized fertility preservation strategies. Integrating genetic markers into clinical practice could revolutionize decision making in fertility preservation for female cancer survivors, enhancing reproductive potential preservation. Background: Recent advances in cancer diagnosis and treatment have significantly improved survival rates among women of reproductive age facing cancer. However, the potential iatrogenic loss of fertility caused by chemotherapeutic agents underscores the need to understand and predict chemotherapy-induced ovarian damage. This study addresses this gap by systematically reviewing the literature to investigate genetic markers associated with chemotherapy-induced ovarian failure (CIOF). Objective: The primary objective is to identify genetic markers linked to CIOF, contributing to a comprehensive understanding of the factors influencing fertility preservation in female cancer survivors. Methods: A systematic review was conducted using PubMed, EMBASE, Web of Science, Scopus, and OVID electronic databases from inception through December 2023. Studies were included if they featured genomic assessments of genes or polymorphisms related to CIOF in women with histologically confirmed tumors. Exclusion criteria comprised in vitro and animal studies, reviews, and pilot studies. The resulting four human-based studies were scrutinized for insights into genetic influences on CIOF. Results: Of the 5179 articles initially identified, four studies met the inclusion criteria, focusing on alkylating agents, particularly cyclophosphamide, and anthracyclines. Su et al. explored CYP3A41B variants, revealing modified associations with CIOF based on age. Charo et al. investigated GSTA1 and CYP2C19 polymorphisms, emphasizing the need to consider age and tamoxifen therapy in assessing associations. Oktay et al. delved into the impact of BRCA mutations on anti-Müllerian hormone (AMH) levels post-chemotherapy, supported by in vitro assays. Van der Perk et al. focused on childhood cancer survivors and revealed significant associations of CYP3A43 and CYP2B6*2 SNPs with AMH levels. Conclusions: This systematic review analyzes evidence regarding genetic markers influencing CIOF, emphasizing the complex interplay of age, specific genetic variants, and chemotherapy regimens. The findings underscore the need for a personalized approach in assessing CIOF risk, integrating genetic markers with traditional ovarian reserve testing. The implications of this study extend to potential advancements in fertility preservation strategies, offering clinicians a comprehensive baseline assessment for tailored interventions based on each patient's unique genetic profile. Further research is essential to validate these findings and establish a robust framework for integrating genetic markers into clinical practice. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
40. A Review on DNA Damaging Patterns and Repair Mechanisms.
- Author
-
Bilal, Muhammad, Maqbool, Tayyaba, Wasim, Muhammad, Tariq, Wajeeha, and Parveen, Bushra
- Subjects
- *
DNA repair , *EXCISION repair , *DNA damage , *ALKYLATING agents , *ONCOLOGIC surgery , *ULTRAVIOLET radiation - Abstract
Various factors and agents cause mutations and damage to DNA, such as UV radiations, alkylating agents, toxins, aromatic compounds, environmental stress and other ways, including spontaneous base DNA damage. These DNA damages could prove deleterious if left un-repaired, leading to cancer and many other unhealthy conditions. As DNA is the genomic material, repairing the damage is more critically important. However, cells have various mechanisms to cope with the damaged DNA, including DNA polymerase self-correction and proofreading, direct reversal of the chemical changes, readily repairing of double helix, base and nucleotide excision repair and mismatch repairs. This review briefly explains DNA damage and repair mechanisms and the disease states associated with them. [ABSTRACT FROM AUTHOR]
- Published
- 2024
41. Lack of mismatch repair enhances resistance to methylating agents for cells deficient in oxidative demethylation.
- Author
-
Gutierrez, Roberto, Chan, Annie Yin S., Seigmund Wai Tsuen Lai, Itoh, Shunsuke, Dong-Hyun Lee, Sun, Kelani, Battad, Alana, Shiuan Chen, O’Connor, Timothy R., and Shuck, Sarah C.
- Subjects
- *
REPLICATION fork , *ALKYLATING agents , *TEMOZOLOMIDE , *ALKYLATION , *PROTEIN deficiency , *DEMETHYLATION - Abstract
The human alkylation B (AlkB) homologs, ALKBH2 and ALKBH3, respond to methylation damage to maintain genomic integrity and cellular viability. Both ALKBH2 and ALKBH3 are direct reversal repair enzymes that remove 1-methyladenine (1meA) and 3-methylcytosine (3meC) lesions commonly generated by alkylating chemotherapeutic agents. Thus, the existence of deficiencies in ALKBH proteins can be exploited in synergy with chemotherapy. In this study, we investigated possible interactions between ALKBH2 and ALKBH3 with other proteins that could alter damage response and discovered an interaction with the mismatch repair (MMR) system. To test whether the lack of active MMR impacts ALKBH2 and/or ALKBH3 response to methylating agents, we generated cells deficient in ALKBH2, ALKBH3, or both in addition to Mlh homolog 1 (MLH1), another MMR protein. We found that MLH1koALKBH3ko cells showed enhanced resistance toward SN1- and SN2-type methylating agents, whereas MLH1koALKBH2ko cells were only resistant to SN1-type methylating agents. Concomitant loss of ALKBH2 and ALKBH3 (ALKBH2ko3ko) rendered cells sensitive to SN1- and SN2-agents, but the additional loss of MLH1 enhanced resistance to both types of damage. We also showed that ALKBH2ko3ko cells have an ATR-dependent arrest at the G2/M checkpoint, increased apoptotic signaling, and replication fork stress in response to methylation. However, these responses were not observed with the loss of functional MLH1 in MLH1koALKBH2ko3ko cells. Finally, in MLH1koALKBH2ko3ko cells, we observed elevated mutant frequency in untreated and temozolomide treated cells. These results suggest that obtaining a more accurate prognosis of chemotherapeutic outcome requires information on the functionality of ALKBH2, ALKBH3, and MLH1. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
42. Substrate-Controlled Regioselective Alkylation of 4-Hydroxycoumarin with Diazo Compounds through TfOH Catalysis.
- Author
-
Xia, Zhimin, Li, Chunyan, Luo, Mengxiang, Gu, Shuangxi, Yan, Qiongjiao, Lv, Jian, Zeng, Jie, and Wang, Haifeng
- Subjects
- *
ALKYLATING agents , *DIAZO compounds , *HETEROCYCLIC chemistry , *NUCLEOPHILES , *CATALYSIS - Abstract
Control of the regioselectivity of nucleophiles toward alkylating agents is a fundamental problem in heterocyclic chemistry. Substrate-controlled TfOH-catalyzed alkylation of 4-hydroxycoumarin with diazo compounds has been developed to provide rapid access to the C3- or O-alkylated 4-hydroxycoumarins with high efficiency. The regioselectivity can be determined by varying the electronic nature of the diazo substrates, especially those bearing a methylthio group. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
43. Menstrual Blood Stem Cells-Derived Exosomes as Promising Therapeutic Tools in Premature Ovarian Insufficiency Induced by Gonadotoxic Systemic Anticancer Treatment.
- Author
-
Robalo Cordeiro, Mariana, Roque, Ricardo, Laranjeiro, Bárbara, Carvalhos, Carlota, and Figueiredo-Dias, Margarida
- Subjects
- *
PREMATURE ovarian failure , *OVARIAN follicle , *GRANULOSA cells , *ALKYLATING agents , *EXTRACELLULAR vesicles - Abstract
Gonadotoxicity resulting from systemic and locoregional cancer treatments significantly threatens women's reproductive health, often culminating in premature ovarian insufficiency. These therapies, particularly alkylating agents and ionizing radiation, induce DNA damage and apoptosis in ovarian follicles, leading to infertility, amenorrhea, and estrogen deficiency, which exacerbate risks of osteoporosis and cardiovascular diseases. Existing fertility preservation methods do not prevent immediate ovarian damage, underscoring the need for innovative protective strategies. Menstrual blood-derived stem cells (MenSC) and their extracellular vesicles (EV) present promising regenerative potential due to their therapeutic cargo delivery and pathway modulation capabilities. Preclinical studies demonstrate that MenSC-derived EV ameliorate premature ovarian insufficiency by inhibiting granulosa cell apoptosis, promoting angiogenesis, and activating pivotal pathways such as SMAD3/AKT/MDM2/P53. However, comprehensive research is imperative to ensure the safety, efficacy, and long-term effects of MenSC-derived EV in clinical practice. In this review, we update the current knowledge and research regarding the use of MenSC-derived EV as a novel therapeutic weapon for ovarian regeneration in the context of gonadotoxicity induced by systemic anticancer treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
44. Accidental Encounter of Repair Intermediates in Alkylated DNA May Lead to Double-Strand Breaks in Resting Cells.
- Author
-
Fujii, Shingo and Fuchs, Robert P.
- Subjects
- *
DNA alkylation , *DOUBLE-strand DNA breaks , *EXCISION repair , *ALKYLATING agents , *CELL proliferation , *DNA damage , *DNA repair - Abstract
In clinics, chemotherapy is often combined with surgery and radiation to increase the chances of curing cancers. In the case of glioblastoma (GBM), patients are treated with a combination of radiotherapy and TMZ over several weeks. Despite its common use, the mechanism of action of the alkylating agent TMZ has not been well understood when it comes to its cytotoxic effects in tumor cells that are mostly non-dividing. The cellular response to alkylating DNA damage is operated by an intricate protein network involving multiple DNA repair pathways and numerous checkpoint proteins that are dependent on the type of DNA lesion, the cell type, and the cellular proliferation state. Among the various alkylating damages, researchers have placed a special on O6-methylguanine (O6-mG). Indeed, this lesion is efficiently removed via direct reversal by O6-methylguanine-DNA methyltransferase (MGMT). As the level of MGMT expression was found to be directly correlated with TMZ efficiency, O6-mG was identified as the critical lesion for TMZ mode of action. Initially, the mode of action of TMZ was proposed as follows: when left on the genome, O6-mG lesions form O6-mG: T mispairs during replication as T is preferentially mis-inserted across O6-mG. These O6-mG: T mispairs are recognized and tentatively repaired by a post-replicative mismatched DNA correction system (i.e., the MMR system). There are two models (futile cycle and direct signaling models) to account for the cytotoxic effects of the O6-mG lesions, both depending upon the functional MMR system in replicating cells. Alternatively, to explain the cytotoxic effects of alkylating agents in non-replicating cells, we have proposed a "repair accident model" whose molecular mechanism is dependent upon crosstalk between the MMR and the base excision repair (BER) systems. The accidental encounter between these two repair systems will cause the formation of cytotoxic DNA double-strand breaks (DSBs). In this review, we summarize these non-exclusive models to explain the cytotoxic effects of alkylating agents and discuss potential strategies to improve the clinical use of alkylating agents. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
45. The impact of treatment for childhood classical Hodgkin lymphoma according to the EuroNet-PHL-C2 protocol on serum anti-Müllerian Hormone.
- Author
-
Drechsel, K C E, Broer, S L, Stoutjesdijk, F S, Broeder, E van Dulmen-den, Beishuizen, A, Wallace, W H, Körholz, D, Mauz-Körholz, C, Hasenclever, D, Cepelova, M, Uyttebroeck, A, Ronceray, L, Twisk, J W R, Kaspers, G J L, and Veening, M A
- Subjects
- *
ANTI-Mullerian hormone , *ALKYLATING agents , *INDUCTION chemotherapy , *HODGKIN'S disease , *MENSTRUAL cycle , *AMENORRHEA - Abstract
STUDY QUESTION What is the impact of the EuroNet-PHL-C2 treatment protocol for children with classical Hodgkin lymphoma (cHL) on gonadal function in girls, based on assessment of serum anti-Müllerian hormone (AMH)? SUMMARY ANSWER Serum AMH levels decreased after induction chemotherapy and increased during subsequent treatment and 2 years of follow-up, with lowest levels in patients treated for advanced stage cHL. WHAT IS KNOWN ALREADY Treatment for cHL, particularly alkylating agents and pelvic irradiation, can be gonadotoxic and result in premature reduction of primordial follicles in females. The current EuroNet-PHL-C2 trial aims to reduce the use of radiotherapy in standard childhood cHL treatment, by intensifying chemotherapy. This study aims to assess the gonadotoxic effect of the EuroNet-PHL-C2 protocol. STUDY DESIGN, SIZE, DURATION This international, prospective, multicenter cohort study is embedded in the EuroNet-PHL-C2 trial, an European phase-3 treatment study evaluating the efficacy of standard cHL treatment with OEPA-COPDAC-28 (OEPA: vincristine, etoposide, prednisone, and doxorubicin; COPDAC-28: cyclophosphamide, vincristine, prednisone, and dacarbazine) versus intensified OEPA-DECOPDAC-21 (DECOPDAC-21: COPDAC with additional doxorubicin and etoposide and 25% more cyclophosphamide) in a randomized setting. Participants were recruited between January 2017 and September 2021. PARTICIPANTS/MATERIALS, SETTING, METHODS Female patients aged ≤18 years, treated according to the EuroNet-PHL-C2 protocol for cHL were recruited across 18 sites in the Netherlands, Belgium, Germany, Austria, and Czech Republic. All parents and patients (aged ≥12 years old) provided written informed consent. Serum AMH levels and menstrual cycle characteristics were evaluated over time (at diagnosis, one to three times during treatment and 2 up to 5 years post-diagnosis) and compared between treatment-levels (TL1, TL2, and TL3) and treatment-arms (OEPA-COPDAC-28 and OEPA-DECOPDAC-21). Serum samples obtained from patients after receiving pelvic radiotherapy were excluded from the main analyses. MAIN RESULTS AND THE ROLE OF CHANCE A total of 104 females, with median age at diagnosis of 15.6 years (IQR 13.7; 17.0), were included in the analysis. Ninety-nine were (post)pubertal. Eighteen girls were diagnosed with an early stage of cHL (TL1) and 86 with intermediate or advanced stage disease (50 TL2 and 36 TL3, 66% received COPDAC-28 and 34% DECOPDAC-21). Five patients received pelvic radiotherapy. Median AMH level at diagnosis was 1.7 µg/l (IQR 0.9; 2.7). After two courses of OEPA chemotherapy, AMH levels decreased substantially in all patients (98% <0.5 µg/l), followed by a significant increase during the consolidation treatment and follow-up. After 2 years, 68% of patients reached their baseline AMH value, with overall median recovery of 129% (IQR 75.0; 208.9) compared to baseline measurement. Five patients (7%) had AMH <0.5 µg/l. In patients treated for advanced stage disease, AMH levels remained significantly lower compared to early- or intermediate stage disease, with median serum AMH of 1.3 µg/l (IQR 0.8; 2.1) after 2 years. Patients who received DECOPDAC-21 consolidation had lower AMH levels during treatment than patients receiving COPDAC-28, but the difference was no longer statistically significant at 2 years post-diagnosis. Of the 35 postmenarchal girls who did not receive hormonal co-treatment, 19 (54%) experienced treatment-induced amenorrhea, two girls had persisting amenorrhea after 2 years. LIMITATIONS, REASONS FOR CAUTION The studied population comprises young girls with diagnosis of cHL often concurring with pubertal transition, during which AMH levels naturally rise. There was no control population, while the interpretation of AMH as a biomarker during childhood is complex. The state of cHL disease may affect AMH levels at diagnosis, potentially complicating assessment of AMH recovery as a comparison with baseline AMH. The current analysis included data up to 2–5 years post-diagnosis. WIDER IMPLICATIONS OF THE FINDINGS The current PANCARE guideline advises to use the cyclophosphamide-equivalent dose score (CED-score, as an estimation of cumulative alkylating agent exposure) with a cut-off of 6000 mg/m2 to identify females aged <25 years at high risk of infertility. All treatment-arms of the EuroNet-PHL-C2 protocol remain below this cut-off, and based on this guideline, girls treated for cHL should therefore be considered low-risk of infertility. However, although we observed an increase in AMH after chemotherapy, it should be noted that not all girls recovered to pre-treatment AMH levels, particularly those treated for advanced stages of cHL. It remains unclear how our measurements relate to age-specific expected AMH levels and patterns. Additional (long-term) data are needed to explore clinical reproductive outcomes of survivors treated according to the EuroNet-PHL-C2 protocol. STUDY FUNDING/COMPETING INTEREST(S) The fertility add-on study was funded by the Dutch charity foundation KiKa (project 257) that funds research on all forms of childhood cancer. C.M-K. D.K. W.H.W. D.H. M.C. A.U. and A.B. were involved in the development of the EuroNet-PHL-C2 regimen. The other authors indicated no potential conflicts of interest. TRIAL REGISTRATION NUMBER N/A. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
46. Pre-mating exposure with hesperidin protects N-ethyl-N-nitrosourea-induced neurotoxicity and congenital abnormalities in next generation of mice as a model of glioma.
- Author
-
Khezri, Saleh, Azizian, Sepideh, and Salimi, Ahmad
- Abstract
Chemical carcinogen-induced oxidative stress has a key role in cell signaling linked to the development of cancer. Oxidative stress leads to oxidative damage to cellular membranes, proteins, chromosomes and genetic material. It is thought that compounds like hesperidin with high antioxidant and anticancer potential can reduce development of cancer induced by chemical carcinogens via neutralizing their oxidative damages. We investigated protective effect of hesperidin against N-Ethyl-N-Nitrosourea (ENU)-induced neurotoxicity, congenital abnormalities and possible brain cancer after exposure of mice during pregnancy as model of glioma. The mice were divided to four groups; control (normal saline), ENU (40 mg/kg daily for three consecutive days from the 17th to the 19th of pregnancy), hesperidin (pretreated with 25 mg/kg for 30 consecutive days, before mating) + ENU and hesperidin alone. Developmental toxicity parameters (the number of pregnant mice, stillbirths, abortion, live and dead offspring), behavioral tests (novel object recognition, open field and elevated plus maze) were performed. Moreover, the activity of butrylcholinesterase and acetylcholinesterase enzymes, oxidative markers and histopathological abnormalities were detected in brain tissue. Our data showed that conversely, the pretreatment of hesperidin reduces various degrees of developmental toxicity, neurobehavioral dysfunction, neurotoxicity, oxidative stress and histopathological abnormalities induced by ENU as a neurotoxic and carcinogenic agent in the next generation. In conclusion, pre-mating exposure with hesperidin may open new avenues for prevention of primary brain cancer in next generation and could be valuable for enhancing the antioxidant defense and minimizing the developmental and neurotoxicity of DNA alkylating agents. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
47. Epithelial–mesenchymal transition in chemoradiation‐induced lung damage: Mechanisms and potential treatment approaches.
- Author
-
Saadh, Mohamed J., Sharma, Pawan, Naser, Israa Habeeb, Kumar, Abhishek, Ravi Kumar, M., Rasulova, Irodakhon, Mohammed, Faraj, Allela, Omer Qutaiba B., Mohammed, Wathiq Kh., Ahmed, Nahed Mahmood, Al‐Ani, Ahmed Muzahem, and Redhee, Ahmed Huseen
- Subjects
ALKYLATING agents ,ANTINEOPLASTIC agents ,CANCER treatment ,OXIDATIVE stress ,BLEOMYCIN - Abstract
Pulmonary injury is one of the key restricting factors for the therapy of malignancies with chemotherapy or following radiotherapy for chest cancers. The lung is a sensitive organ to some severely toxic antitumor drugs, consisting of bleomycin and alkylating agents. Furthermore, treatment with radiotherapy may drive acute and late adverse impacts on the lung. The major consequences of radiotherapy and chemotherapy in the lung are pneumonitis and fibrosis. Pneumonitis may arise some months to a few years behind cancer therapy. However, fibrosis is a long‐term effect that appears years after chemo/or radiotherapy. Several mechanisms such as oxidative stress and severe immune reactions are implicated in the progression of pulmonary fibrosis. Epithelial–mesenchymal transition (EMT) is offered as a pivotal mechanism for lung fibrosis behind chemotherapy and radiotherapy. It seems that pulmonary fibrosis is the main consequence of EMT after chemo/radiotherapy. Several biological processes, consisting of the liberation of pro‐inflammatory and pro‐fibrosis molecules, oxidative stress, upregulation of nuclear factor of κB and Akt, epigenetic changes, and some others, may participate in EMT and pulmonary fibrosis behind cancer therapy. In this review, we aim to discuss how chemotherapy or radiotherapy may promote EMT and lung fibrosis. Furthermore, we review potential targets and effective agents to suppress EMT and lung fibrosis after cancer therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
48. Manganese‐Catalyzed α‐Alkylation of Sulfones using Alcohols via a Hydrogen‐Borrowing Strategy: Synthesis of Branched Sulfones.
- Author
-
Verma, Ashutosh and Elias, Anil J.
- Subjects
MANGANESE catalysts ,ALKYLATING agents ,BENZYL alcohol ,SULFONES ,BIOCHEMICAL substrates - Abstract
Herein, we report an efficient and sustainable manganese‐catalyzed α‐C−H bond alkylation methodology to synthesize branched sulfones via a hydrogen borrowing pathway. The air‐stable and phosphine‐free Mn‐catalyst, (NNN)Mn(II)Cl2 was synthesized by using an earth‐abundant, commercially available, and inexpensive precursor MnCl2.4H2O, and a stable NNN pincer i. e. [N‐((benzimidazole‐2‐yl)methyl)quinoline‐8‐amine] ligand system. Taking benzyl phenyl sulfones as substrates, and benzyl alcohol derivatives as alkylating agents, a range of branched sulfones were synthesized in 40–82 % isolated yields using (NNN)Mn(II)Cl2 complex as the catalyst under open‐air conditions. Control experiments and deuterium incorporation studies have also been conducted to investigate the possible reaction mechanism and to provide evidence for the hydrogen borrowing pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
49. Loncastuximab Tesirine in the Treatment of Relapsed or Refractory Diffuse Large B-Cell Lymphoma.
- Author
-
Juárez-Salcedo, Luis Miguel, Nimkar, Santosh, Corazón, Ana María, and Dalia, Samir
- Subjects
- *
DIFFUSE large B-cell lymphomas , *RITUXIMAB , *B cells , *HISTOCOMPATIBILITY antigens , *STEM cell transplantation , *MYC oncogenes , *CHIMERIC antigen receptors , *ALKYLATING agents , *ANTIGEN receptors - Abstract
Currently, a significant percentage of patients with DLBCL are refractory or relapse after a first line of immunochemotherapy. Second relapses after autologous stem cell transplantation or chimeric antigen receptor T-cell therapies present few treatment options and do not yield good results. New molecules have entered the immunotherapy arsenal. Loncastuximab tesirine comprises a humanized anti-CD19 monoclonal conjugated antibody, which consists of an anti-CD19 antibody and cytotoxic alkylating agent, SG3199. Several studies have proven its efficacy in the treatment of refractory cases of DLBCL with a good safety profile, with the main adverse effects being neutropenia, thrombopenia, and liver enzyme involvement. In this review, we explain the mechanism of action of this molecule, the clinical data that have led to its acceptance by the FDA, and the new therapeutic options that are proposed in association with this drug. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
50. An efficient synthesis of O- and N- alkyl derivatives of 4-aminobenzoic acid and evaluation of their anticancer properties.
- Author
-
Hasan, Erum, Ali, Syed Nawazish, Bano, Zarina, Begum, Sabira, Ali, Sundus, Shams, Afshan, and Siddiqui, Bina S.
- Subjects
PROTON magnetic resonance ,ALKYLATING agents ,ONE-way analysis of variance ,ACID derivatives ,BENZOIC acid ,NUCLEAR magnetic resonance - Abstract
A series of twenty alkyl derivatives (2–21) of 4-amino benzoic acid (1, PABA) have been prepared using potassium carbonate and opportune alkylating agents under simple and mild reaction conditions. Compounds (16–21) are reported for the first time. Electron impact mass spectrometry (EIMS), Fourier transform infrared (FTIR) and Proton nuclear magnetic resonance (
1 H-NMR) spectroscopic techniques were adopted for the characterization of these analogues. In the present study, the cytotoxic screening of sixteen compounds (3, 5–11, 13 and 15–21) was also achieved against lung (NCI-H460) and oral squamous carcinoma (CAL-27) cell lines. Compound 20 has shown magnificent inhibitory properties against NCI-H460 cell line (IC50 15.59 and 20.04 µM, respectively) at a lower dose than that of the control (cisplatin; IC50 21.00 µM). One-way analysis of variance (ANOVA), t-test and Pearson correlation coefficient (PCC) have been performed to determine the reliability of current data through statistical package for the social sciences (SPSS). [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.