File Download

There are no files associated with this item.

  Links for fulltext
     (May Require Subscription)
Supplementary

Article: Pharmacological inhibition of serine synthesis enhances temozolomide efficacy by decreasing O6-methylguanine DNA methyltransferase (MGMT) expression and reactive oxygen species (ROS)-mediated DNA damage in glioblastoma

TitlePharmacological inhibition of serine synthesis enhances temozolomide efficacy by decreasing O6-methylguanine DNA methyltransferase (MGMT) expression and reactive oxygen species (ROS)-mediated DNA damage in glioblastoma
Authors
Issue Date2022
PublisherNature Publishing Group. The Journal's web site is located at https://www.nature.com/labinvest/
Citation
Laboratory Investigation, 2022, v. 102 n. 2, p. 194-203 How to Cite?
AbstractGlioblastoma (GBM) is the most malignant primary tumor in the central nervous system of adults. Temozolomide (TMZ), an alkylating agent, is the first-line chemotherapeutic agent for GBM patients. However, its efficacy is often limited by innate or acquired chemoresistance. Cancer cells can rewire their metabolic programming to support rapid growth and sustain cell survival against chemotherapies. An example is the de novo serine synthesis pathway (SSP), one of the main branches from glycolysis that is highly activated in multiple cancers in promoting cancer progression and inducing chemotherapy resistance. However, the roles of SSP in TMZ therapy for GBM patients remain unexplored. In this study, we employed NCT503, a highly selective inhibitor of phosphoglycerate dehydrogenase (PHGDH, the first rate-limiting enzyme of SSP), to study whether inhibition of SSP may enhance TMZ efficacy in MGMT-positive GBMs. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), flowcytometry and colony formation assays demonstrated that NCT503 worked synergistically with TMZ in suppressing GBM cell growth and inducing apoptosis in T98G and U118 cells in vitro. U118 and patient-derived GBM subcutaneous xenograft models showed that combined NCT503 and TMZ treatment inhibited GBM growth and promoted apoptosis more significantly than would each treatment alone in vivo. Mechanistically, we found that NCT503 treatment decreased MGMT expression possibly by modulating the Wnt/β-catenin pathway. Moreover, intracellular levels of reactive oxygen species were elevated especially when NCT503 and TMZ treatments were combined, and the synergistic effects could be partially negated by NAC, a classic scavenger of reactive oxygen species. Taken together, these results suggest that NCT503 may be a promising agent for augmenting TMZ efficacy in the treatment of GBM, especially in TMZ-resistant GBMs with high expression of MGMT.
Persistent Identifierhttp://hdl.handle.net/10722/307728
ISSN
2021 Impact Factor: 5.502
2020 SCImago Journal Rankings: 1.542
ISI Accession Number ID

 

DC FieldValueLanguage
dc.contributor.authorJin, L-
dc.contributor.authorKiang, KMY-
dc.contributor.authorCheng, SY-
dc.contributor.authorLeung, GKK-
dc.date.accessioned2021-11-12T13:36:57Z-
dc.date.available2021-11-12T13:36:57Z-
dc.date.issued2022-
dc.identifier.citationLaboratory Investigation, 2022, v. 102 n. 2, p. 194-203-
dc.identifier.issn0023-6837-
dc.identifier.urihttp://hdl.handle.net/10722/307728-
dc.description.abstractGlioblastoma (GBM) is the most malignant primary tumor in the central nervous system of adults. Temozolomide (TMZ), an alkylating agent, is the first-line chemotherapeutic agent for GBM patients. However, its efficacy is often limited by innate or acquired chemoresistance. Cancer cells can rewire their metabolic programming to support rapid growth and sustain cell survival against chemotherapies. An example is the de novo serine synthesis pathway (SSP), one of the main branches from glycolysis that is highly activated in multiple cancers in promoting cancer progression and inducing chemotherapy resistance. However, the roles of SSP in TMZ therapy for GBM patients remain unexplored. In this study, we employed NCT503, a highly selective inhibitor of phosphoglycerate dehydrogenase (PHGDH, the first rate-limiting enzyme of SSP), to study whether inhibition of SSP may enhance TMZ efficacy in MGMT-positive GBMs. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), flowcytometry and colony formation assays demonstrated that NCT503 worked synergistically with TMZ in suppressing GBM cell growth and inducing apoptosis in T98G and U118 cells in vitro. U118 and patient-derived GBM subcutaneous xenograft models showed that combined NCT503 and TMZ treatment inhibited GBM growth and promoted apoptosis more significantly than would each treatment alone in vivo. Mechanistically, we found that NCT503 treatment decreased MGMT expression possibly by modulating the Wnt/β-catenin pathway. Moreover, intracellular levels of reactive oxygen species were elevated especially when NCT503 and TMZ treatments were combined, and the synergistic effects could be partially negated by NAC, a classic scavenger of reactive oxygen species. Taken together, these results suggest that NCT503 may be a promising agent for augmenting TMZ efficacy in the treatment of GBM, especially in TMZ-resistant GBMs with high expression of MGMT.-
dc.languageeng-
dc.publisherNature Publishing Group. The Journal's web site is located at https://www.nature.com/labinvest/-
dc.relation.ispartofLaboratory Investigation-
dc.titlePharmacological inhibition of serine synthesis enhances temozolomide efficacy by decreasing O6-methylguanine DNA methyltransferase (MGMT) expression and reactive oxygen species (ROS)-mediated DNA damage in glioblastoma-
dc.typeArticle-
dc.identifier.emailKiang, KMY: mykiang@hku.hk-
dc.identifier.emailCheng, SY: yincyns@hku.hk-
dc.identifier.emailLeung, GKK: gkkleung@hku.hk-
dc.identifier.authorityLeung, GKK=rp00522-
dc.description.naturelink_to_subscribed_fulltext-
dc.identifier.doi10.1038/s41374-021-00666-7-
dc.identifier.pmid34625658-
dc.identifier.scopuseid_2-s2.0-85116576839-
dc.identifier.hkuros329847-
dc.identifier.volume102-
dc.identifier.issue2-
dc.identifier.spage194-
dc.identifier.epage203-
dc.identifier.isiWOS:000705700100001-
dc.publisher.placeUnited Kingdom-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats