Drug (ID: DG00020) and It's Reported Resistant Information
Name
Carmustine
Synonyms
Carmustine; carmustine; 154-93-8; 1,3-Bis(2-chloroethyl)-1-nitrosourea; BCNU; Carmustin; Nitrumon; Carmubris; Gliadel; BiCNU; Bi CNU; Carmustinum; Bischlorethylnitrosurea; Bischlorethylnitrosourea; Carmustina; Becenun; Becenum; Bischloroethyl nitrosourea; N,N'-BIS(2-CHLOROETHYL)-N-NITROSOUREA; Bis(2-chloroethyl)nitrosourea; Urea, N,N'-bis(2-chloroethyl)-N-nitroso-; Gliadel Wafer; FDA 0345; Bischloroethylnitrosourea; SRI 1720; 1,3-Bis(2-chloroethyl)nitrosourea; BiCNU (TN); Carmustinum [INN-Latin]; Carmustina [INN-Spanish]; DTI 015; NCI-C04773; SK; BCNU; Injectable carmustine, Direct Therapeutics
    Click to Show/Hide
Indication
In total 2 Indication(s)
Brain cancer [ICD-11: 2A00]
Approved
[1]
Liver cancer [ICD-11: 2C12]
Approved
[1]
Structure
Drug Resistance Disease(s)
Disease(s) with Clinically Reported Resistance for This Drug (1 diseases)
Brain cancer [ICD-11: 2A00]
[2]
Disease(s) with Resistance Information Discovered by Cell Line Test for This Drug (1 diseases)
Brain cancer [ICD-11: 2A00]
[3]
Target Human Deoxyribonucleic acid (hDNA) NOUNIPROTAC [1]
Click to Show/Hide the Molecular Information and External Link(s) of This Drug
Formula
C5H9Cl2N3O2
IsoSMILES
C(CCl)NC(=O)N(CCCl)N=O
InChI
1S/C5H9Cl2N3O2/c6-1-3-8-5(11)10(9-12)4-2-7/h1-4H2,(H,8,11)
InChIKey
DLGOEMSEDOSKAD-UHFFFAOYSA-N
PubChem CID
2578
ChEBI ID
CHEBI:3423
TTD Drug ID
D01OXI
INTEDE ID
DR0278
DrugBank ID
DB00262
Type(s) of Resistant Mechanism of This Drug
  DISM: Drug Inactivation by Structure Modification
  EADR: Epigenetic Alteration of DNA, RNA or Protein
  IDUE: Irregularity in Drug Uptake and Drug Efflux
  UAPP: Unusual Activation of Pro-survival Pathway
Drug Resistance Data Categorized by Their Corresponding Diseases
ICD-02: Benign/in-situ/malignant neoplasm
Click to Show/Hide the Resistance Disease of This Class
Brain cancer [ICD-11: 2A00]
Click to Show/Hide
Drug Resistance Data Categorized by Their Corresponding Mechanisms
       Drug Inactivation by Structure Modification (DISM) Click to Show/Hide
Key Molecule: Glutathione S-transferase P (GSTP1) [2]
Molecule Alteration Expression
Up-regulation
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue N.A.
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
EDR assay
Mechanism Description In vitro drug resistance in malignant gliomas was independent of prior therapy. High-grade glioblastomas showed a lower level of extreme drug resistance than low-grade astrocytomas to cisplatin (11% versus 27%), temozolomide (14% versus 27%), irinotecan (33% versus 53%), and BCNU (29% versus 38%). A substantial percentage of brain tumors overexpressed biomarkers associated with drug resistance, including MGMT (67%), GSTP1 (49%), and mutant p53 (41%). MGMT and GSTP1 overexpression was independently associated with in vitro resistance to BCNU, whereas coexpression of these two markers was associated with the greatest degree of BCNU resistance.
Key Molecule: Glutathione S-transferase P (GSTP1) [2]
Molecule Alteration Expression
Up-regulation
Resistant Disease Anaplastic astrocytoma [ICD-11: 2A00.04]
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue N.A.
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
Oncotech EDR assay
Mechanism Description GSTP1 is the first major mechanism of resistance alkylator agents encounter after entering the cancer cell cytoplasm. GSTP1 acts to enzymatically conjugate glutathione to the reactive metabolites of BCNU. The mechanisms by which GSTP1 may be up-regulated in gliomas are under investigation. Constitutive expression is thought to be influenced by the proximal promoter factor Sp1, whereas increased expression levels may result from stabilization of GSTP1 mRNA. GSTP1 expression has been reported to be induced by drug exposure, indicating that it may play a role in acquired drug resistance.
       Epigenetic Alteration of DNA, RNA or Protein (EADR) Click to Show/Hide
Key Molecule: hsa-mir-21 [1]
Molecule Alteration Expression
Up-regulation
Resistant Disease Glioma [ICD-11: 2A00.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SWOZ2 cells Brain Homo sapiens (Human) N.A.
SWOZ2-BCNU cells Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
RT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR21 enhanced glioma cells resistance to carmustine via decreasing Spry2 expression.
Key Molecule: hsa-mir-221 [3]
Molecule Alteration Expression
Up-regulation
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
PI3-k/PTEN/AKT signaling axis Activation hsa04151
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
In Vivo Model Nude mouse xenograft model Mus musculus
Experiment for
Molecule Alteration
qRT-PCR
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-221 regulated cell proliferation and BCNU resistance in glioma cells. Overexpression of miR-221 led to cell survival and BCNU resistance and reduced cell apoptosis induced by BCNU, whereas knockdown of miR-221 inhibited cell proliferation and prompted BCNU sensitivity and cell apoptosis. Further investigation revealed that miR-221 down-regulated PTEN and activated Akt, which resulted in cell survival and BCNU resistance. Overexpression of PTEN lacking 3'UTR or PI3-k/Akt specific inhibitor wortmannin attenuated miR-221-mediated BCNU resistance and prompted cell apoptosis. We propose that miR-221 regulated cell proliferation and BCNU resistance in glioma cells by targeting PI3-k/PTEN/Akt signaling axis.
       Irregularity in Drug Uptake and Drug Efflux (IDUE) Click to Show/Hide
Key Molecule: Multidrug resistance protein 1 (ABCB1) [2]
Molecule Alteration Expression
Up-regulation
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue N.A.
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
EDR assay
Mechanism Description In vitro drug resistance in malignant gliomas was independent of prior therapy. High-grade glioblastomas showed a lower level of extreme drug resistance than low-grade astrocytomas to cisplatin (11% versus 27%), temozolomide (14% versus 27%), irinotecan (33% versus 53%), and BCNU (29% versus 38%). A substantial percentage of brain tumors overexpressed biomarkers associated with drug resistance, including MGMT (67%), GSTP1 (49%), and mutant p53 (41%). MGMT and GSTP1 overexpression was independently associated with in vitro resistance to BCNU, whereas coexpression of these two markers was associated with the greatest degree of BCNU resistance.
       Unusual Activation of Pro-survival Pathway (UAPP) Click to Show/Hide
Key Molecule: Protein sprouty homolog 2 (SPRY2) [1]
Molecule Alteration Expression
Down-regulation
Resistant Disease Glioma [ICD-11: 2A00.1]
Experimental Note Revealed Based on the Cell Line Data
In Vitro Model SWOZ2 cells Brain Homo sapiens (Human) N.A.
SWOZ2-BCNU cells Brain Homo sapiens (Human) N.A.
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR21 enhanced glioma cells resistance to carmustine via decreasing Spry2 expression.
Key Molecule: Phosphatase and tensin homolog (PTEN) [3]
Molecule Alteration Expression
Down-regulation
Resistant Disease Glioblastoma [ICD-11: 2A00.02]
Experimental Note Revealed Based on the Cell Line Data
Cell Pathway Regulation Cell apoptosis Inhibition hsa04210
Cell proliferation Activation hsa05200
PI3K/PTEN/AKT signaling axis Activation hsa04151
In Vitro Model U251 cells Brain Homo sapiens (Human) CVCL_0021
U87 cells Brain Homo sapiens (Human) CVCL_0022
Experiment for
Molecule Alteration
Western blot analysis
Experiment for
Drug Resistance
CCK8 assay
Mechanism Description miR-221 regulated cell proliferation and BCNU resistance in glioma cells. Overexpression of miR-221 led to cell survival and BCNU resistance and reduced cell apoptosis induced by BCNU, whereas knockdown of miR-221 inhibited cell proliferation and prompted BCNU sensitivity and cell apoptosis. Further investigation revealed that miR-221 down-regulated PTEN and activated Akt, which resulted in cell survival and BCNU resistance. Overexpression of PTEN lacking 3'UTR or PI3-k/Akt specific inhibitor wortmannin attenuated miR-221-mediated BCNU resistance and prompted cell apoptosis. We propose that miR-221 regulated cell proliferation and BCNU resistance in glioma cells by targeting PI3-k/PTEN/Akt signaling axis.
Key Molecule: Methylated-DNA--protein-cysteine methyltransferase (MGMT) [2]
Molecule Alteration Expression
Up-regulation
Resistant Disease Malignant glioma [ICD-11: 2A00.2]
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue N.A.
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
EDR assay
Mechanism Description In vitro drug resistance in malignant gliomas was independent of prior therapy. High-grade glioblastomas showed a lower level of extreme drug resistance than low-grade astrocytomas to cisplatin (11% versus 27%), temozolomide (14% versus 27%), irinotecan (33% versus 53%), and BCNU (29% versus 38%). A substantial percentage of brain tumors overexpressed biomarkers associated with drug resistance, including MGMT (67%), GSTP1 (49%), and mutant p53 (41%). MGMT and GSTP1 overexpression was independently associated with in vitro resistance to BCNU, whereas coexpression of these two markers was associated with the greatest degree of BCNU resistance.
Key Molecule: Methylated-DNA--protein-cysteine methyltransferase (MGMT) [2]
Molecule Alteration Expression
Up-regulation
Resistant Disease Anaplastic astrocytoma [ICD-11: 2A00.04]
Experimental Note Identified from the Human Clinical Data
In Vitro Model Malignant gliomas tissue N.A.
Experiment for
Molecule Alteration
Immunohistochemistry assay
Experiment for
Drug Resistance
Oncotech EDR assay
Mechanism Description For drugs that have evaded cytosolic mechanisms of drug resistance, the nucleus is equipped with the capacity to remove BCNU or temozolomide alkyl groups from the O6-position of guanine via a reaction catalyzed by MGMT. Repair occurs before cross-link formation and involves an irreversible stoichiometric covalent transfer of the O6-alkyl DNA adduct to a cysteine within the active site of MGMT, resulting in the inactivation and subsequent depletion of enzyme activity. MGMT-mediated repair is rapid, with a half-life of 35 hours. MGMT enzyme recovery occurs via de novo synthesis. In malignant glioma patients, MGMT overexpression has been associated with resistance to BCNU and similar alkylating agents and was an independent predictor of poor survival. MGMT is also thought to contribute to temozolomide resistance, which we did not detect in our study. This may be related to the in vitro pharmacokinetic differences between BCNU and temozolomide.
References
Ref 1 MiR-21 enhanced glioma cells resistance to carmustine via decreasing Spry2 expression. Eur Rev Med Pharmacol Sci. 2017 Nov;21(22):5065-5071. doi: 10.26355/eurrev_201711_13819.
Ref 2 In vitro drug response and molecular markers associated with drug resistance in malignant gliomas .Clin Cancer Res. 2006 Aug 1;12(15):4523-32. doi: 10.1158/1078-0432.CCR-05-1830. 10.1158/1078-0432.CCR-05-1830
Ref 3 MicroRNA-221 targeting PI3-K/Akt signaling axis induces cell proliferation and BCNU resistance in human glioblastoma. Neuropathology. 2014 Oct;34(5):455-64. doi: 10.1111/neup.12129. Epub 2014 Apr 30.

If you find any error in data or bug in web service, please kindly report it to Dr. Sun and Dr. Zhang.