Disease Information
General Information of the Disease (ID: DIS00050)
Name |
Chronic myeloid leukemia
|
---|---|
ICD |
ICD-11: 2A20
|
Resistance Map |
Type(s) of Resistant Mechanism of This Disease
Drug Resistance Data Categorized by Drug
Approved Drug(s)
16 drug(s) in total
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: hsa-mir-21 | [1] | |||
Sensitive Disease | Chronic myelogenous leukemia [ICD-11: 2A20.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Arsenic trioxide | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell growth | Inhibition | hsa05200 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR-21 post-transcriptionally down-regulates tumor suppressor PDCD4. AMO-miR-21 sensitized leukemic k562 cells to ATO by inducing apoptosis partially due to its up-regulation of PDCD4 protein level. | |||
|
||||
Key Molecule: Programmed cell death protein 4 (PDCD4) | [1] | |||
Sensitive Disease | Chronic myelogenous leukemia [ICD-11: 2A20.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Arsenic trioxide | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell growth | Inhibition | hsa05200 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR-21 post-transcriptionally down-regulates tumor suppressor PDCD4. AMO-miR-21 sensitized leukemic k562 cells to ATO by inducing apoptosis partially due to its up-regulation of PDCD4 protein level. |
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [2] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F359V |
||
Resistant Drug | Bosutinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Overall survival assay; Event-free survival (EFS) assay | |||
Mechanism Description | Patients with more than one BCR-ABL1 mutation fare worse than those with no or one mutation. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [2] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.M351T |
||
Resistant Drug | Bosutinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Overall survival assay; Event-free survival (EFS) assay | |||
Mechanism Description | Patients with more than one BCR-ABL1 mutation fare worse than those with no or one mutation. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [3] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F359I |
||
Resistant Drug | Bosutinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Tritiated thymidine incorporation assay | |||
Mechanism Description | L248R was identified in a patient with lymphoid Blast Crisis (BC) CML (Patient no. 1), in cis with a pre-existing mutation. The patient initially presented with an imatinib-resistant F359I mutation. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [3] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.L248R |
||
Resistant Drug | Bosutinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Tritiated thymidine incorporation assay | |||
Mechanism Description | Mutations may impair TkI activity by directly or indirectly impairing the drug binding to the protein. We report the discovery of three new BCR/ABL mutations, L248R, T315V, and F317R identified in two patients with CML (L248R and T315V) and in one patient with Ph+ acute lymphoblastic leukemia (ALL) (F317R). | |||
|
||||
Key Molecule: GTPase Nras (NRAS) | [4], [5] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.G12V |
||
Resistant Drug | Bosutinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | JAKT2/STAT signaling pathway | Activation | hsa04030 | |
RAF/KRAS/MEK signaling pathway | Activation | hsa04010 | ||
In Vitro Model | HL60 cells | Peripheral blood | Homo sapiens (Human) | CVCL_0002 |
U937 cells | Blood | Homo sapiens (Human) | CVCL_0007 | |
K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 | |
KCL-22 cells | Bone marrow | Homo sapiens (Human) | CVCL_2091 | |
Sup-B15 cells | Bone marrow | Homo sapiens (Human) | CVCL_0103 | |
HEL cells | Blood | Homo sapiens (Human) | CVCL_0001 | |
HMC-1.2 cells | Blood | Homo sapiens (Human) | CVCL_H205 | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Next-generation sequencing assay; Sanger Sequencing assay | |||
Mechanism Description | This mutation is well known for its effects on proliferation and its association with AML and MPN, suggesting that this variant might have been involved in the TkI resistance of this patient. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: Tyrosine-protein kinase JAK2 (JAK3) | [6] | |||
Sensitive Disease | Primary myelofibrosis [ICD-11: 2A20.2] | |||
Molecule Alteration | Function | Inhibition |
||
Sensitive Drug | Danazol | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | In non-transplant candidates, conventional treatment for anemia includes androgens, prednisone, thalidomide, and danazol; for symptomatic splenomegaly, hydroxyurea and ruxolitinib; and for constitutional symptoms, ruxolitinib. Fedratinib, another JAK2 inhibitor, has now been FDA-approved for use in ruxolitinib failures. |
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [7], [8], [9] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F317L |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Progression-free survival assay; Overall survival assay | |||
Mechanism Description | After 13 months of therapy a progression of disease to accelerated phase was detected and a second mutational screening analysis performed at that time revealed the absence of M244 V and the appearance of M351T mutation. After 14 months of therapy, a third mutational analysis was performed which revealed the disappearance of M351T mutation and the acquisition of a new F317L mutation. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [2], [9], [10] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.Y253H |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.V338F |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [8], [9], [12] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.V299L |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.V268A |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [2], [9], [10] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.T315I |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [3], [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.T315A |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.Q252H |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.M351T |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [13], [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.M244V |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [13], [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.L387M |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.L384M |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.L298V |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.L248V |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.H396R |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [8], [9], [10] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.G250E |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [13], [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F359V |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F359C |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F317V |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F317I |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F317C |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F311L |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E459K |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [9], [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E355G |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence 23223358. We confirmed the high frequency of SFks involvement in Tyrosine kinase inhibitor-resistant CML (52% of the cases) and even further in progressive disease and blast crises (60% of the cases). The SFks deregulation is also observed in patients harboring BCR-ABL mutations. In T315I and F317L mutated patients, CML-resistance appears to be promoted by SFks kinase protein reactivation once the BCR-ABL mutated clone has decreased on Omacetaxine. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E255V |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E255K |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.D325G |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [8] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.T495R |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Mechanism Description | The most common mechanism of acquired resistance in CML in imatinib era is the acquisition of BCR-ABL kinase domain mutations with decreased sensitivity to the drug. Our findings demonstrate the potential hazards of sequential kinase inhibitor therapy and suggest a role for a combination of ABL kinase inhibitors, perhaps including drugs with different mechanisms of action, to prevent the outgrowth of cells harboring drug-resistant BCR-ABL mutations. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [13] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.M388L |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
RNA sequencing assay | |||
Mechanism Description | The presence of BCR-ABL oncogene mutations in patients with chronic myeloid leukemia (CML) may be responsible for the failure of tyrosine kinase inhibitor (TkI) treatment. In addition to 9 point mutations (G250E / F317L, F359V, L387M, Y253H, M388L, M244V, T315I, D276G), 35 bp insertion between exons 8 and 9 and deletion exon 7 were detected. Our results demonstrate that direct sequencing is suitable for routine clinical monitoring patients with CML and may be useful for optimizing therapy. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [13] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.D276G |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
RNA sequencing assay | |||
Mechanism Description | The presence of BCR-ABL oncogene mutations in patients with chronic myeloid leukemia (CML) may be responsible for the failure of tyrosine kinase inhibitor (TkI) treatment. In addition to 9 point mutations (G250E / F317L, F359V, L387M, Y253H, M388L, M244V, T315I, D276G), 35 bp insertion between exons 8 and 9 and deletion exon 7 were detected. Our results demonstrate that direct sequencing is suitable for routine clinical monitoring patients with CML and may be useful for optimizing therapy. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [9] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.Y353H |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | We confirmed the high frequency of SFks involvement in Tyrosine kinase inhibitor-resistant CML (52% of the cases) and even further in progressive disease and blast crises (60% of the cases). The SFks deregulation is also observed in patients harboring BCR-ABL mutations. In T315I and F317L mutated patients, CML-resistance appears to be promoted by SFks kinase protein reactivation once the BCR-ABL mutated clone has decreased on Omacetaxine. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [9] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.Y253F |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | We confirmed the high frequency of SFks involvement in Tyrosine kinase inhibitor-resistant CML (52% of the cases) and even further in progressive disease and blast crises (60% of the cases). The SFks deregulation is also observed in patients harboring BCR-ABL mutations. In T315I and F317L mutated patients, CML-resistance appears to be promoted by SFks kinase protein reactivation once the BCR-ABL mutated clone has decreased on Omacetaxine. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [9] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.V379I |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | We confirmed the high frequency of SFks involvement in Tyrosine kinase inhibitor-resistant CML (52% of the cases) and even further in progressive disease and blast crises (60% of the cases). The SFks deregulation is also observed in patients harboring BCR-ABL mutations. In T315I and F317L mutated patients, CML-resistance appears to be promoted by SFks kinase protein reactivation once the BCR-ABL mutated clone has decreased on Omacetaxine. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [9] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.L273M |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | We confirmed the high frequency of SFks involvement in Tyrosine kinase inhibitor-resistant CML (52% of the cases) and even further in progressive disease and blast crises (60% of the cases). The SFks deregulation is also observed in patients harboring BCR-ABL mutations. In T315I and F317L mutated patients, CML-resistance appears to be promoted by SFks kinase protein reactivation once the BCR-ABL mutated clone has decreased on Omacetaxine. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.V299L |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Circulating-free DNA assay; Whole exome sequencing assay | |||
Mechanism Description | In patients treated sequentially with dasatinib, nilotinib, or both TkIs after imatinib failure who had developed resistance to second-line treatment, analysis of the individual components of the compound mutations revealed that the identities of component mutations reflected the type of prior drug exposure. Therefore, in all patients treated with dasatinib, at least 1 component of the compound mutations was V299L, F317L, or E255k, all of which have been reported in clinical or in vitro resistance to dasatinib. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F317L |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Circulating-free DNA assay; Whole exome sequencing assay | |||
Mechanism Description | In patients treated sequentially with dasatinib, nilotinib, or both TkIs after imatinib failure who had developed resistance to second-line treatment, analysis of the individual components of the compound mutations revealed that the identities of component mutations reflected the type of prior drug exposure. Therefore, in all patients treated with dasatinib, at least 1 component of the compound mutations was V299L, F317L, or E255k, all of which have been reported in clinical or in vitro resistance to dasatinib. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E255K |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Circulating-free DNA assay; Whole exome sequencing assay | |||
Mechanism Description | In patients treated sequentially with dasatinib, nilotinib, or both TkIs after imatinib failure who had developed resistance to second-line treatment, analysis of the individual components of the compound mutations revealed that the identities of component mutations reflected the type of prior drug exposure. Therefore, in all patients treated with dasatinib, at least 1 component of the compound mutations was V299L, F317L, or E255k, all of which have been reported in clinical or in vitro resistance to dasatinib. | |||
|
||||
Key Molecule: hsa_circ_BA9.3 | [14] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Ku812 cells | Bone marrow | Homo sapiens (Human) | CVCL_0379 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCk reagent assay; Flow cytometry assay | |||
Mechanism Description | CircBA9.3 promoted cell proliferation and reduced the sensitivity of leukaemic cells to TkIs through up-regulation of the ABL1 and BCR-ABL1 protein expression levels. | |||
Key Molecule: hsa-miR-29a-3p | [15] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Lin-CD34+CD38- cells | Bone | Homo sapiens (Human) | N.A. |
Lin-CD34-CD38- CML cells | Bone | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Annexin V assay | |||
Mechanism Description | The up-regulation of miR29a-3p observed in CML LSCs led to the down-regulation of its target TET2 and conferred TkI-resistance to CML LSCs in vitro. | |||
Key Molecule: hsa-miR-494-3p | [15] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Lin-CD34+CD38- cells | Bone | Homo sapiens (Human) | N.A. |
Lin-CD34-CD38- CML cells | Bone | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Annexin V assay | |||
Mechanism Description | miR494-3p down-regulation in CML LSCs, leading to c-MYC up-regulation, was able to decrease TkI-induced apoptosis. | |||
Key Molecule: hsa-miR-660-5p | [15] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Lin-CD34+CD38- cells | Bone | Homo sapiens (Human) | N.A. |
Lin-CD34-CD38- CML cells | Bone | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Annexin V assay | |||
Mechanism Description | The up-regulation of miR660-5p observed in CML LSCs led to the down-regulation of its target EPAS1 and conferred TkI-resistance to CML LSCs in vitro. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [9] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.D444Y |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | We confirmed the high frequency of SFks involvement in Tyrosine kinase inhibitor-resistant CML (52% of the cases) and even further in progressive disease and blast crises (60% of the cases). The SFks deregulation is also observed in patients harboring BCR-ABL mutations. In T315I and F317L mutated patients, CML-resistance appears to be promoted by SFks kinase protein reactivation once the BCR-ABL mutated clone has decreased on Omacetaxine. | |||
|
||||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [14] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Ku812 cells | Bone marrow | Homo sapiens (Human) | CVCL_0379 | |
Experiment for Molecule Alteration |
Western blot analysis; qRT-PCR | |||
Experiment for Drug Resistance |
CCk reagent assay; Flow cytometry assay | |||
Mechanism Description | CircBA9.3 promoted cell proliferation and reduced the sensitivity of leukaemic cells to TkIs through up-regulation of the ABL1 and BCR-ABL1 protein expression levels. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [14] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Ku812 cells | Bone marrow | Homo sapiens (Human) | CVCL_0379 | |
Experiment for Molecule Alteration |
Western blot analysis; qRT-PCR | |||
Experiment for Drug Resistance |
CCk reagent assay; Flow cytometry assay | |||
Mechanism Description | CircBA9.3 promoted cell proliferation and reduced the sensitivity of leukaemic cells to TkIs through up-regulation of the ABL1 and BCR-ABL1 protein expression levels. | |||
Key Molecule: Myc proto-oncogene protein (MYC) | [15] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Lin-CD34+CD38- cells | Bone | Homo sapiens (Human) | N.A. |
Lin-CD34-CD38- CML cells | Bone | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Annexin V assay | |||
Mechanism Description | miR494-3p down-regulation in CML LSCs, leading to c-MYC up-regulation, was able to decrease TkI-induced apoptosis. | |||
Key Molecule: Hypoxia-inducible factor 2-alpha (EPAS1) | [15] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Lin-CD34+CD38- cells | Bone | Homo sapiens (Human) | N.A. |
Lin-CD34-CD38- CML cells | Bone | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Annexin V assay | |||
Mechanism Description | The up-regulation of miR660-5p observed in CML LSCs led to the down-regulation of its target EPAS1 and conferred TkI-resistance to CML LSCs in vitro. | |||
Key Molecule: Methylcytosine dioxygenase TET2 (TET2) | [15] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Lin-CD34+CD38- cells | Bone | Homo sapiens (Human) | N.A. |
Lin-CD34-CD38- CML cells | Bone | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Annexin V assay | |||
Mechanism Description | The up-regulation of miR29a-3p observed in CML LSCs led to the down-regulation of its target TET2 and conferred TkI-resistance to CML LSCs in vitro. | |||
Key Molecule: GTPase Nras (NRAS) | [4], [5] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.G12V |
||
Resistant Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | JAKT2/STAT signaling pathway | Activation | hsa04030 | |
RAF/KRAS/MEK signaling pathway | Activation | hsa04010 | ||
In Vitro Model | HL60 cells | Peripheral blood | Homo sapiens (Human) | CVCL_0002 |
U937 cells | Blood | Homo sapiens (Human) | CVCL_0007 | |
K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 | |
KCL-22 cells | Bone marrow | Homo sapiens (Human) | CVCL_2091 | |
Sup-B15 cells | Bone marrow | Homo sapiens (Human) | CVCL_0103 | |
HEL cells | Blood | Homo sapiens (Human) | CVCL_0001 | |
HMC-1.2 cells | Blood | Homo sapiens (Human) | CVCL_H205 | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Next-generation sequencing assay; Sanger Sequencing assay | |||
Mechanism Description | This mutation is well known for its effects on proliferation and its association with AML and MPN, suggesting that this variant might have been involved in the TkI resistance of this patient. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: hsa-mir-217 | [16] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Dasatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell viability | Inhibition | hsa05200 | ||
miR217/AGR2 signaling pathway | Regulation | hsa05206 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Ku812 cells | Bone marrow | Homo sapiens (Human) | CVCL_0379 | |
kCL22 cells | Pleural effusion | Homo sapiens (Human) | CVCL_2091 | |
In Vivo Model | NRG mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay | |||
Mechanism Description | miR-217 sensitizes chronic myelogenous leukemia cells to tyrosine kinase inhibitors by downregulating pro-oncogenic anterior gradient 2. | |||
Key Molecule: hsa-mir-217 | [17] | |||
Sensitive Disease | Chronic myelogenous Ph(+) leukemia [ICD-11: 2A20.1] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Bcr/Abl-expressing k562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
K562DR cells | Blood | Homo sapiens (Human) | CVCL_4V59 | |
K562NR cells | Blood | Homo sapiens (Human) | CVCL_4V63 | |
In Vivo Model | BALB/c nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Forced expression of miR-217 inhibited expression of DNMT3A through a miR-217-binding site within the 3'-untranslated region of DNMT3A and sensitized these cells to growth inhibition mediated by the TkI. long-term exposure of CML k562 cells to ABL TkI such as dasatinib and nilotinib decreased the levels of miR-217 and increased the levels of DNMT1 and DNMT3A, as well as resulting in acquisition of TkI resistance. | |||
|
||||
Key Molecule: Anterior gradient protein 2 homolog (AGR2) | [16] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Dasatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell viability | Inhibition | hsa05200 | ||
miR217/AGR2 signaling pathway | Regulation | hsa05206 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Ku812 cells | Bone marrow | Homo sapiens (Human) | CVCL_0379 | |
kCL22 cells | Pleural effusion | Homo sapiens (Human) | CVCL_2091 | |
In Vivo Model | NRG mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay | |||
Mechanism Description | miR-217 sensitizes chronic myelogenous leukemia cells to tyrosine kinase inhibitors by downregulating pro-oncogenic anterior gradient 2. | |||
Key Molecule: DNA (cytosine-5)-methyltransferase 3A (DNMT3A) | [17] | |||
Sensitive Disease | Chronic myelogenous Ph(+) leukemia [ICD-11: 2A20.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Dasatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Bcr/Abl-expressing k562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
K562DR cells | Blood | Homo sapiens (Human) | CVCL_4V59 | |
K562NR cells | Blood | Homo sapiens (Human) | CVCL_4V63 | |
In Vivo Model | BALB/c nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Forced expression of miR-217 inhibited expression of DNMT3A through a miR-217-binding site within the 3'-untranslated region of DNMT3A and sensitized these cells to growth inhibition mediated by the TkI. long-term exposure of CML k562 cells to ABL TkI such as dasatinib and nilotinib decreased the levels of miR-217 and increased the levels of DNMT1 and DNMT3A, as well as resulting in acquisition of TkI resistance. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: Multidrug resistance protein 1 (ABCB1) | [18] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Daunorubicin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | NCI-H460 cells | Lung | Homo sapiens (Human) | CVCL_0459 |
K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 | |
HEK293 cells | Kidney | Homo sapiens (Human) | CVCL_0045 | |
K562-R cells | Pleural effusion | Homo sapiens (Human) | CVCL_5950 | |
NCI-H460/VBL cells | Bone marrow | Homo sapiens (Human) | CVCL_0459 | |
In Vivo Model | SCID beige mice | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | In ABCB1-overexpressing cell lines, HG-829 significantly enhanced cytotoxicity to daunorubicin, paclitaxel, vinblastine, vincristine, and etoposide. Coadministration of HG-829 fully restored in vivo antitumor activity of daunorubicin in mice without added toxicity. Functional assays showed that HG-829 is not a Pgp substrate or competitive inhibitor of Pgp-mediated drug efflux but rather acts as a noncompetitive modulator of P-glycoprotein transport function. |
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: hsa-mir-21 | [19] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Doxorubicin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
PI3K/AKT/mTOR signaling pathway | Activation | hsa04151 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
K562/A02 cells | Blood | Homo sapiens (Human) | CVCL_0368 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR-21 is associated with inactivation of PTEN, a know tumor suppressor gene, resulting in activation of PI3k/Akt/mTOR signaling pathway, Akt promotes cell survival by inhibiting apoptosis through its ability to phosphorylate/inactivate downstream targets of apoptotic machinery. ADR sensitivity is associated with up-regulation of PTEN resulting from the inhibition of miR-21 expression. | |||
|
||||
Key Molecule: Phosphatase and tensin homolog (PTEN) | [19] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Doxorubicin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
PI3K/AKT/mTOR signaling pathway | Activation | hsa04151 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
K562/A02 cells | Blood | Homo sapiens (Human) | CVCL_0368 | |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR-21 is associated with inactivation of PTEN, a know tumor suppressor gene, resulting in activation of PI3k/Akt/mTOR signaling pathway, Akt promotes cell survival by inhibiting apoptosis through its ability to phosphorylate/inactivate downstream targets of apoptotic machinery. ADR sensitivity is associated with up-regulation of PTEN resulting from the inhibition of miR-21 expression. | |||
Key Molecule: Annexin A1 (ANXA1) | [20] | |||
Resistant Disease | Chronic myelogenous leukemia [ICD-11: 2A20.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Doxorubicin | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | The downregulated ANXA1,whose new role in apoptosis and cancer revealed recently,expression contributes considerably to the observed drug resistance in k562/ADR cells. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: hsa-mir-9 | [21] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Doxorubicin | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR9 regulates the multidrug resistance of chronic myelogenous leukemia by targeting ABCB1. | |||
Key Molecule: hsa-mir-181c | [22] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Doxorubicin | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
PI3K/AKT signaling pathway | Inhibition | hsa04151 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Ku812 cells | Bone marrow | Homo sapiens (Human) | CVCL_0379 | |
kCL22 cells | Pleural effusion | Homo sapiens (Human) | CVCL_2091 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | miR-181c directly targeted and inhibited the ST8SIA4 expression, as well as miR-181c was inversely correlated with the levels of ST8SIA4 expression in CML cell lines and samples. Moreover, ST8SIA4 could reverse the effect of miR-181c on drug resistance in k562 and k562/ADR cells in vitro. Upregulation of miR-181c sensitized k562/ADR cells to adriamycin in vivo through directly suppressing ST8SIA4 expression. Further investigation showed that miR-181c mediated the activity of phosphoinositide-3 kinase (PI3k)/AkT signal pathway, and inhibition of PI3k/Akt in k562 cells counteracted miR-181c-mediated MDR phenotype. | |||
|
||||
Key Molecule: Multidrug resistance protein 1 (ABCB1) | [21] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Doxorubicin | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | miR9 regulates the multidrug resistance of chronic myelogenous leukemia by targeting ABCB1. | |||
|
||||
Key Molecule: Sialyltransferase St8Sia IV (SIAT8D) | [22] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Doxorubicin | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
PI3K/AKT signaling pathway | Inhibition | hsa04151 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Ku812 cells | Bone marrow | Homo sapiens (Human) | CVCL_0379 | |
kCL22 cells | Pleural effusion | Homo sapiens (Human) | CVCL_2091 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | miR-181c directly targeted and inhibited the ST8SIA4 expression, as well as miR-181c was inversely correlated with the levels of ST8SIA4 expression in CML cell lines and samples. Moreover, ST8SIA4 could reverse the effect of miR-181c on drug resistance in k562 and k562/ADR cells in vitro. Upregulation of miR-181c sensitized k562/ADR cells to adriamycin in vivo through directly suppressing ST8SIA4 expression. Further investigation showed that miR-181c mediated the activity of phosphoinositide-3 kinase (PI3k)/AkT signal pathway, and inhibition of PI3k/Akt in k562 cells counteracted miR-181c-mediated MDR phenotype. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: Tyrosine-protein kinase JAK2 (JAK3) | [6] | |||
Sensitive Disease | Primary myelofibrosis [ICD-11: 2A20.2] | |||
Molecule Alteration | Function | Inhibition |
||
Sensitive Drug | Fedratinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | In non-transplant candidates, conventional treatment for anemia includes androgens, prednisone, thalidomide, and danazol; for symptomatic splenomegaly, hydroxyurea and ruxolitinib; and for constitutional symptoms, ruxolitinib. Fedratinib, another JAK2 inhibitor, has now been FDA-approved for use in ruxolitinib failures. | |||
Key Molecule: Tyrosine-protein kinase JAK2 (JAK3) | [23] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.V617F (c.1849G>T) |
||
Sensitive Drug | Fedratinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HEL cells | Blood | Homo sapiens (Human) | CVCL_0001 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
XTT assay | |||
Mechanism Description | The missense mutation p.V617F (c.1849G>T) in gene JAK2 cause the sensitivity of Fedratinib by aberration of the drug's therapeutic target |
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: hsa-mir-29a | [24] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Fludarabine | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | p53 signaling pathway | Inhibition | hsa04115 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | miR-29a can activate p53 and induce apoptosis in a p53-dependent manner. |
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [25] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.Y253H+p.F317L |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | Among the 32 patients with baseline mutation, mutations including M244V, G250E, E255k, M351T, H396R, S417Y, E450k and E459k disappeared in 8 patients and new mutations were detected in 9 patients, all of which were T315I. Among the 23 patients without baseline mutation, 4 patients showed newly developed mutations including T315I, T315I + E459k, M244V and F359V. The T315I was the most frequently detected mutation in imatinib therapy (16%, 9 of 55) as well as in dasatinib or nilotinib therapy (24%, 11 of 44). Patients with imatinib resistant baseline mutations had a higher rate of mutation development during dasatinib or nilotinib treatment compared to patients without baseline mutations (28% vs. 17%). | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [25] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.T315I+p.E459K |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | Among the 32 patients with baseline mutation, mutations including M244V, G250E, E255k, M351T, H396R, S417Y, E450k and E459k disappeared in 8 patients and new mutations were detected in 9 patients, all of which were T315I. Among the 23 patients without baseline mutation, 4 patients showed newly developed mutations including T315I, T315I + E459k, M244V and F359V. The T315I was the most frequently detected mutation in imatinib therapy (16%, 9 of 55) as well as in dasatinib or nilotinib therapy (24%, 11 of 44). Patients with imatinib resistant baseline mutations had a higher rate of mutation development during dasatinib or nilotinib treatment compared to patients without baseline mutations (28% vs. 17%). | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [25] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.P480L |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | Among the 32 patients with baseline mutation, mutations including M244V, G250E, E255k, M351T, H396R, S417Y, E450k and E459k disappeared in 8 patients and new mutations were detected in 9 patients, all of which were T315I. Among the 23 patients without baseline mutation, 4 patients showed newly developed mutations including T315I, T315I + E459k, M244V and F359V. The T315I was the most frequently detected mutation in imatinib therapy (16%, 9 of 55) as well as in dasatinib or nilotinib therapy (24%, 11 of 44). Patients with imatinib resistant baseline mutations had a higher rate of mutation development during dasatinib or nilotinib treatment compared to patients without baseline mutations (28% vs. 17%). | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [25] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.M244V+p.G250E |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | Among the 32 patients with baseline mutation, mutations including M244V, G250E, E255k, M351T, H396R, S417Y, E450k and E459k disappeared in 8 patients and new mutations were detected in 9 patients, all of which were T315I. Among the 23 patients without baseline mutation, 4 patients showed newly developed mutations including T315I, T315I + E459k, M244V and F359V. The T315I was the most frequently detected mutation in imatinib therapy (16%, 9 of 55) as well as in dasatinib or nilotinib therapy (24%, 11 of 44). Patients with imatinib resistant baseline mutations had a higher rate of mutation development during dasatinib or nilotinib treatment compared to patients without baseline mutations (28% vs. 17%). | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [26], [27], [28] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.G250E |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | Among the 32 patients with baseline mutation, mutations including M244V, G250E, E255k, M351T, H396R, S417Y, E450k and E459k disappeared in 8 patients and new mutations were detected in 9 patients, all of which were T315I. Among the 23 patients without baseline mutation, 4 patients showed newly developed mutations including T315I, T315I + E459k, M244V and F359V. The T315I was the most frequently detected mutation in imatinib therapy (16%, 9 of 55) as well as in dasatinib or nilotinib therapy (24%, 11 of 44). Patients with imatinib resistant baseline mutations had a higher rate of mutation development during dasatinib or nilotinib treatment compared to patients without baseline mutations (28% vs. 17%). | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [27], [28], [29] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F359V |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | Among the 32 patients with baseline mutation, mutations including M244V, G250E, E255k, M351T, H396R, S417Y, E450k and E459k disappeared in 8 patients and new mutations were detected in 9 patients, all of which were T315I. Among the 23 patients without baseline mutation, 4 patients showed newly developed mutations including T315I, T315I + E459k, M244V and F359V. The T315I was the most frequently detected mutation in imatinib therapy (16%, 9 of 55) as well as in dasatinib or nilotinib therapy (24%, 11 of 44). Patients with imatinib resistant baseline mutations had a higher rate of mutation development during dasatinib or nilotinib treatment compared to patients without baseline mutations (28% vs. 17%). | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [29], [30], [31] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E459K |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | Among the 32 patients with baseline mutation, mutations including M244V, G250E, E255k, M351T, H396R, S417Y, E450k and E459k disappeared in 8 patients and new mutations were detected in 9 patients, all of which were T315I. Among the 23 patients without baseline mutation, 4 patients showed newly developed mutations including T315I, T315I + E459k, M244V and F359V. The T315I was the most frequently detected mutation in imatinib therapy (16%, 9 of 55) as well as in dasatinib or nilotinib therapy (24%, 11 of 44). Patients with imatinib resistant baseline mutations had a higher rate of mutation development during dasatinib or nilotinib treatment compared to patients without baseline mutations (28% vs. 17%). | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [25] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E450K |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | Among the 32 patients with baseline mutation, mutations including M244V, G250E, E255k, M351T, H396R, S417Y, E450k and E459k disappeared in 8 patients and new mutations were detected in 9 patients, all of which were T315I. Among the 23 patients without baseline mutation, 4 patients showed newly developed mutations including T315I, T315I + E459k, M244V and F359V. The T315I was the most frequently detected mutation in imatinib therapy (16%, 9 of 55) as well as in dasatinib or nilotinib therapy (24%, 11 of 44). Patients with imatinib resistant baseline mutations had a higher rate of mutation development during dasatinib or nilotinib treatment compared to patients without baseline mutations (28% vs. 17%). | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [25] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E255K+p.T315I |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | Among the 32 patients with baseline mutation, mutations including M244V, G250E, E255k, M351T, H396R, S417Y, E450k and E459k disappeared in 8 patients and new mutations were detected in 9 patients, all of which were T315I. Among the 23 patients without baseline mutation, 4 patients showed newly developed mutations including T315I, T315I + E459k, M244V and F359V. The T315I was the most frequently detected mutation in imatinib therapy (16%, 9 of 55) as well as in dasatinib or nilotinib therapy (24%, 11 of 44). Patients with imatinib resistant baseline mutations had a higher rate of mutation development during dasatinib or nilotinib treatment compared to patients without baseline mutations (28% vs. 17%). | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [26], [27], [32] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E255K |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | Among the 32 patients with baseline mutation, mutations including M244V, G250E, E255k, M351T, H396R, S417Y, E450k and E459k disappeared in 8 patients and new mutations were detected in 9 patients, all of which were T315I. Among the 23 patients without baseline mutation, 4 patients showed newly developed mutations including T315I, T315I + E459k, M244V and F359V. The T315I was the most frequently detected mutation in imatinib therapy (16%, 9 of 55) as well as in dasatinib or nilotinib therapy (24%, 11 of 44). Patients with imatinib resistant baseline mutations had a higher rate of mutation development during dasatinib or nilotinib treatment compared to patients without baseline mutations (28% vs. 17%). | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [33] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.Y320C |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Overall survival assay; Event-free survival (EFS) assay | |||
Mechanism Description | Compared to non-mutated patients, subjects with point mutations had a worse response to dasatinib, with significantly lower rates of complete cytogenetic response (57 vs 32 %), higher percentage of primary resistance (16/36 vs 6/40) and a trend towards a shorter median event-free survival. In elderly patients, the presence of a mutation at the time of imatinib failure is associated with a worse response to dasatinib therapy. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [9], [33], [34] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.V299L |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Overall survival assay; Event-free survival (EFS) assay | |||
Mechanism Description | Compared to non-mutated patients, subjects with point mutations had a worse response to dasatinib, with significantly lower rates of complete cytogenetic response (57 vs 32 %), higher percentage of primary resistance (16/36 vs 6/40) and a trend towards a shorter median event-free survival. In elderly patients, the presence of a mutation at the time of imatinib failure is associated with a worse response to dasatinib therapy. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [33] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.V256L |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Overall survival assay; Event-free survival (EFS) assay | |||
Mechanism Description | Compared to non-mutated patients, subjects with point mutations had a worse response to dasatinib, with significantly lower rates of complete cytogenetic response (57 vs 32 %), higher percentage of primary resistance (16/36 vs 6/40) and a trend towards a shorter median event-free survival. In elderly patients, the presence of a mutation at the time of imatinib failure is associated with a worse response to dasatinib therapy. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [33], [35] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.T277A |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Overall survival assay; Event-free survival (EFS) assay | |||
Mechanism Description | Compared to non-mutated patients, subjects with point mutations had a worse response to dasatinib, with significantly lower rates of complete cytogenetic response (57 vs 32 %), higher percentage of primary resistance (16/36 vs 6/40) and a trend towards a shorter median event-free survival. In elderly patients, the presence of a mutation at the time of imatinib failure is associated with a worse response to dasatinib therapy. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [33] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.S438C |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Overall survival assay; Event-free survival (EFS) assay | |||
Mechanism Description | Compared to non-mutated patients, subjects with point mutations had a worse response to dasatinib, with significantly lower rates of complete cytogenetic response (57 vs 32 %), higher percentage of primary resistance (16/36 vs 6/40) and a trend towards a shorter median event-free survival. In elderly patients, the presence of a mutation at the time of imatinib failure is associated with a worse response to dasatinib therapy. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [33] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.M351K |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Overall survival assay; Event-free survival (EFS) assay | |||
Mechanism Description | Compared to non-mutated patients, subjects with point mutations had a worse response to dasatinib, with significantly lower rates of complete cytogenetic response (57 vs 32 %), higher percentage of primary resistance (16/36 vs 6/40) and a trend towards a shorter median event-free survival. In elderly patients, the presence of a mutation at the time of imatinib failure is associated with a worse response to dasatinib therapy. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [33] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.K378R |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Overall survival assay; Event-free survival (EFS) assay | |||
Mechanism Description | Compared to non-mutated patients, subjects with point mutations had a worse response to dasatinib, with significantly lower rates of complete cytogenetic response (57 vs 32 %), higher percentage of primary resistance (16/36 vs 6/40) and a trend towards a shorter median event-free survival. In elderly patients, the presence of a mutation at the time of imatinib failure is associated with a worse response to dasatinib therapy. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [33] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E494G |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Overall survival assay; Event-free survival (EFS) assay | |||
Mechanism Description | Compared to non-mutated patients, subjects with point mutations had a worse response to dasatinib, with significantly lower rates of complete cytogenetic response (57 vs 32 %), higher percentage of primary resistance (16/36 vs 6/40) and a trend towards a shorter median event-free survival. In elderly patients, the presence of a mutation at the time of imatinib failure is associated with a worse response to dasatinib therapy. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [33], [36], [37] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E450G |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Overall survival assay; Event-free survival (EFS) assay | |||
Mechanism Description | Compared to non-mutated patients, subjects with point mutations had a worse response to dasatinib, with significantly lower rates of complete cytogenetic response (57 vs 32 %), higher percentage of primary resistance (16/36 vs 6/40) and a trend towards a shorter median event-free survival. In elderly patients, the presence of a mutation at the time of imatinib failure is associated with a worse response to dasatinib therapy. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [27], [28], [29] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E355G |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Overall survival assay; Event-free survival (EFS) assay | |||
Mechanism Description | Compared to non-mutated patients, subjects with point mutations had a worse response to dasatinib, with significantly lower rates of complete cytogenetic response (57 vs 32 %), higher percentage of primary resistance (16/36 vs 6/40) and a trend towards a shorter median event-free survival. In elderly patients, the presence of a mutation at the time of imatinib failure is associated with a worse response to dasatinib therapy. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [33] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.A399T |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Overall survival assay; Event-free survival (EFS) assay | |||
Mechanism Description | Compared to non-mutated patients, subjects with point mutations had a worse response to dasatinib, with significantly lower rates of complete cytogenetic response (57 vs 32 %), higher percentage of primary resistance (16/36 vs 6/40) and a trend towards a shorter median event-free survival. In elderly patients, the presence of a mutation at the time of imatinib failure is associated with a worse response to dasatinib therapy. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [38] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.N368S |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Denaturing high performance liquid chromatography (dHPLC) assay; Direct DNA sequencing method assay | |||
Experiment for Drug Resistance |
Overall survival assay | |||
Mechanism Description | Fifteen different types of mutations (T315I, E255k, G250E, M351T, F359C, G251E, Y253H, V289F, E355G, N368S, L387M, H369R, A397P, E355A, D276G), including 2 novel mutations were identified, with T315I as the predominant type of mutation. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [38] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.G251E |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Denaturing high performance liquid chromatography (dHPLC) assay; Direct DNA sequencing method assay | |||
Experiment for Drug Resistance |
Overall survival assay | |||
Mechanism Description | Fifteen different types of mutations (T315I, E255k, G250E, M351T, F359C, G251E, Y253H, V289F, E355G, N368S, L387M, H369R, A397P, E355A, D276G), including 2 novel mutations were identified, with T315I as the predominant type of mutation. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [38], [39], [40] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.A397P |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Denaturing high performance liquid chromatography (dHPLC) assay; Direct DNA sequencing method assay | |||
Experiment for Drug Resistance |
Overall survival assay | |||
Mechanism Description | Fifteen different types of mutations (T315I, E255k, G250E, M351T, F359C, G251E, Y253H, V289F, E355G, N368S, L387M, H369R, A397P, E355A, D276G), including 2 novel mutations were identified, with T315I as the predominant type of mutation. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.V338F |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.V268A |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.T315A |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [31], [39], [40] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.L298V |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [25], [31], [36] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F317V |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F317I |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F317C |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.D325G |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Sanger sequencing assay | |||
Mechanism Description | For CML patients on TkI therapy, 70% of double mutations in the BCR-ABL1 kinase domain detected by direct sequencing are compound mutations. Sequential, branching, and parallel routes to compound mutations were observed, suggesting complex patterns of emergence. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [41] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.Q252K |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Next generation sequencing assay | |||
Experiment for Drug Resistance |
Event-free survival assay; Overall survival assay | |||
Mechanism Description | HSCT is an important salvage option for TkI-resistant patients with or without BCR-ABL1 mutations. Patients with mutations were more likely to develop advanced disease and had worse outcomes after HSCT. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [42] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.Q252M |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Pyrosequencing analysis | |||
Experiment for Drug Resistance |
Progression-free survival assay; Overall survival assay | |||
Mechanism Description | Imatinib resistance in chronic myeloid leukemia (CML) is commonly due to BCR-ABL kinase domain mutations (kDMs). | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [42] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.P310S |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Pyrosequencing analysis | |||
Experiment for Drug Resistance |
Progression-free survival assay; Overall survival assay | |||
Mechanism Description | Imatinib resistance in chronic myeloid leukemia (CML) is commonly due to BCR-ABL kinase domain mutations (kDMs). | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [36], [42] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.H396P |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Pyrosequencing analysis | |||
Experiment for Drug Resistance |
Progression-free survival assay; Overall survival assay | |||
Mechanism Description | Imatinib resistance in chronic myeloid leukemia (CML) is commonly due to BCR-ABL kinase domain mutations (kDMs). | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [26], [27], [28] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F311I |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
PCR-Invader assay; Direct sequencing assay | |||
Experiment for Drug Resistance |
Progression-free survival assay; Overall survival assay | |||
Mechanism Description | The PCR-Invader assay used in this study is an appropriate tool for the screening of mutations during TkI therapy. High Sokal score is only predictive factor for emergence of mutation in CML-CP. P-loop mutations were associated with poor PFS in CML-CP. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [43] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.Q252E |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | In late CP or advanced CML, ABL-kinase mutations occur as an intraclonal event in the primitive Ph1+ stem cell compartments with progression of this clone towards IM-resistant blast phase. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [44] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Structural mutation | Structural variation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
ASO-PCR and sequencing assay | |||
Experiment for Drug Resistance |
Event-free survival assay; Overall survival assay | |||
Mechanism Description | Mutations in the kinase domain (kD) of BCR-ABL are the leading cause of acquired imatinib resistance. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [31], [36], [45] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.L364I |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Real-time Taqman assay; Direct sequencing assay | |||
Experiment for Drug Resistance |
Progression-free survival assay | |||
Mechanism Description | Point mutation was the major mechanism of primary cytogenetic resistance to imatinib mesylate in the present study. Patients with mutations had inferior progression-free survival compared to those without mutations. Resistance is higher among patients with advanced CML. Point mutations in the ABL kinase domain and amplification of the BCR-ABL fusion gene have emerged as important mechanisms responsible for resistance to imatinib. Biochemical and cellular assays have demonstrated that different BCR-ABL mutations might result in varying levels of resistance. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [38], [46], [47] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.V289F |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Overall survival assay | |||
Mechanism Description | Point mutations were detected in 36 of 154 patients by direct sequencing. In our series, the single most common mutations were G250E, E255k/V, and M351T. The presence of mutations correlated significantly with accelerated phase, lack of molecular response, and lower cytogenetic and hematological responses. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [9], [36], [48] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.L273M |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Sanger sequencing assay | |||
Mechanism Description | Ponatinib was highly active in heavily pretreated patients with Ph-positive leukemias with resistance to tyrosine kinase inhibitors, including patients with the BCR-ABL T315I mutation, other mutations, or no mutations. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [49] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.N374Y |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Nested RT-PC assay; Gene sequencing assay | |||
Experiment for Drug Resistance |
Event-free survival assay; Overall survival assay | |||
Mechanism Description | Presence of mutations predicted for poorer responses and EFS to dose escalation. IM dose escalation is likely to be effective only in those harboring no or relatively sensitive kD mutations. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [50] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E453G |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Sanger sequencing assay | |||
Mechanism Description | The data suggest that some BCR-ABL1 mutations may persist at undetectable levels for many years after changing therapy, and can be reselected and confer resistance to subsequent inhibitors. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [50] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E275K |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Sanger sequencing assay | |||
Mechanism Description | The data suggest that some BCR-ABL1 mutations may persist at undetectable levels for many years after changing therapy, and can be reselected and confer resistance to subsequent inhibitors. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [35] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.L340L |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | The frequency of ABL mutations in CML patients resistant to imatinib is high and is more frequent among those with clonal cytogenetic evolution. The change to second-generation TkI can overcome imatinib resistance in most of the mutated patients. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [35] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.D276A |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | The frequency of ABL mutations in CML patients resistant to imatinib is high and is more frequent among those with clonal cytogenetic evolution. The change to second-generation TkI can overcome imatinib resistance in most of the mutated patients. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [29], [30], [31] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.I418V |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | The most frequent mutant is M244V, followed by Y253H, F359C/V/I, G250E, E255k, and T315I. Only seven patients (9%) have T315I mutants, and all showed hematologic resistance. Three of them were in the ECP and three in the LCP. Look-back studies show that mutants were detected 0-20 (median 7) months ahead of the appearance of clinical resistance in 15 tested patients with acquired resistance. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [39] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E453L |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | The most frequent mutant is M244V, followed by Y253H, F359C/V/I, G250E, E255k, and T315I. Only seven patients (9%) have T315I mutants, and all showed hematologic resistance. Three of them were in the ECP and three in the LCP. Look-back studies show that mutants were detected 0-20 (median 7) months ahead of the appearance of clinical resistance in 15 tested patients with acquired resistance. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [37], [39] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E450A |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | The most frequent mutant is M244V, followed by Y253H, F359C/V/I, G250E, E255k, and T315I. Only seven patients (9%) have T315I mutants, and all showed hematologic resistance. Three of them were in the ECP and three in the LCP. Look-back studies show that mutants were detected 0-20 (median 7) months ahead of the appearance of clinical resistance in 15 tested patients with acquired resistance. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [39] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E279Y |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | The most frequent mutant is M244V, followed by Y253H, F359C/V/I, G250E, E255k, and T315I. Only seven patients (9%) have T315I mutants, and all showed hematologic resistance. Three of them were in the ECP and three in the LCP. Look-back studies show that mutants were detected 0-20 (median 7) months ahead of the appearance of clinical resistance in 15 tested patients with acquired resistance. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [51] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.L387F |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Bidirectional DNA sequencing method assay | |||
Experiment for Drug Resistance |
Progression-free survival assay; Overall survival assay | |||
Mechanism Description | This report expands the spectrum of BCR-ABL mutations and stresses the use of mutation testing in imatinib-resistant patients for continuation of treatment procedure. The most commonly mutated region was drug-binding site (29%) followed by P-loop region (26%) and most patients bearing them were in accelerated phase and blastic phase. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [31], [36], [51] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E459G |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Bidirectional DNA sequencing method assay | |||
Experiment for Drug Resistance |
Progression-free survival assay; Overall survival assay | |||
Mechanism Description | This report expands the spectrum of BCR-ABL mutations and stresses the use of mutation testing in imatinib-resistant patients for continuation of treatment procedure. The most commonly mutated region was drug-binding site (29%) followed by P-loop region (26%) and most patients bearing them were in accelerated phase and blastic phase. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [51] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E453A |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Bidirectional DNA sequencing method assay | |||
Experiment for Drug Resistance |
Progression-free survival assay; Overall survival assay | |||
Mechanism Description | This report expands the spectrum of BCR-ABL mutations and stresses the use of mutation testing in imatinib-resistant patients for continuation of treatment procedure. The most commonly mutated region was drug-binding site (29%) followed by P-loop region (26%) and most patients bearing them were in accelerated phase and blastic phase. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [51] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E279A |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Bidirectional DNA sequencing method assay | |||
Experiment for Drug Resistance |
Progression-free survival assay; Overall survival assay | |||
Mechanism Description | This report expands the spectrum of BCR-ABL mutations and stresses the use of mutation testing in imatinib-resistant patients for continuation of treatment procedure. The most commonly mutated region was drug-binding site (29%) followed by P-loop region (26%) and most patients bearing them were in accelerated phase and blastic phase. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [51] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.D276N |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Bidirectional DNA sequencing method assay | |||
Experiment for Drug Resistance |
Progression-free survival assay; Overall survival assay | |||
Mechanism Description | This report expands the spectrum of BCR-ABL mutations and stresses the use of mutation testing in imatinib-resistant patients for continuation of treatment procedure. The most commonly mutated region was drug-binding site (29%) followed by P-loop region (26%) and most patients bearing them were in accelerated phase and blastic phase. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [51] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.S438C |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Bidirectional DNA sequencing method assay | |||
Experiment for Drug Resistance |
Progression-free survival assay; Overall survival assay | |||
Mechanism Description | This report expands the spectrum of BCR-ABL mutations and stresses the use of mutation testing in imatinib-resistant patients for continuation of treatment procedure. The most commonly mutated region was drug-binding site (29%) followed by P-loop region (26%) and most patients bearing them were in accelerated phase and blastic phase. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [25], [29], [36] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.S417Y |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
DNA sequencing assay | |||
Experiment for Drug Resistance |
Progression-free survival assay | |||
Mechanism Description | We conclude that the currently recommended 10-fold threshold to trigger mutation screening is insensitive and not universally applicable. kinase domain mutations predict a shorter progression-free survival. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [36] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.G251D |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
DNA sequencing assay | |||
Experiment for Drug Resistance |
Progression-free survival assay | |||
Mechanism Description | We conclude that the currently recommended 10-fold threshold to trigger mutation screening is insensitive and not universally applicable. kinase domain mutations predict a shorter progression-free survival. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [27], [28], [29] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F382L |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
DNA sequencing assay | |||
Experiment for Drug Resistance |
Progression-free survival assay | |||
Mechanism Description | We conclude that the currently recommended 10-fold threshold to trigger mutation screening is insensitive and not universally applicable. kinase domain mutations predict a shorter progression-free survival. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [31], [36] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E453K |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
DNA sequencing assay | |||
Experiment for Drug Resistance |
Progression-free survival assay | |||
Mechanism Description | We conclude that the currently recommended 10-fold threshold to trigger mutation screening is insensitive and not universally applicable. kinase domain mutations predict a shorter progression-free survival. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [36] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E453D |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
DNA sequencing assay | |||
Experiment for Drug Resistance |
Progression-free survival assay | |||
Mechanism Description | We conclude that the currently recommended 10-fold threshold to trigger mutation screening is insensitive and not universally applicable. kinase domain mutations predict a shorter progression-free survival. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [36] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E352G |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
DNA sequencing assay | |||
Experiment for Drug Resistance |
Progression-free survival assay | |||
Mechanism Description | We conclude that the currently recommended 10-fold threshold to trigger mutation screening is insensitive and not universally applicable. kinase domain mutations predict a shorter progression-free survival. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [36] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E352D |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
DNA sequencing assay | |||
Experiment for Drug Resistance |
Progression-free survival assay | |||
Mechanism Description | We conclude that the currently recommended 10-fold threshold to trigger mutation screening is insensitive and not universally applicable. kinase domain mutations predict a shorter progression-free survival. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [36] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E282G |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
DNA sequencing assay | |||
Experiment for Drug Resistance |
Progression-free survival assay | |||
Mechanism Description | We conclude that the currently recommended 10-fold threshold to trigger mutation screening is insensitive and not universally applicable. kinase domain mutations predict a shorter progression-free survival. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [36] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E279Z |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
DNA sequencing assay | |||
Experiment for Drug Resistance |
Progression-free survival assay | |||
Mechanism Description | We conclude that the currently recommended 10-fold threshold to trigger mutation screening is insensitive and not universally applicable. kinase domain mutations predict a shorter progression-free survival. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [36] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.D482V |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
DNA sequencing assay | |||
Experiment for Drug Resistance |
Progression-free survival assay | |||
Mechanism Description | We conclude that the currently recommended 10-fold threshold to trigger mutation screening is insensitive and not universally applicable. kinase domain mutations predict a shorter progression-free survival. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [52] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.K419E |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | We confirm the previously described poor prognosis of CML patients with mutations in the BCR-ABL1 kD, since 40.0% of our CML patients who harbored a BCR-ABL1 kD mutation died from CML while receiving TkI treatment. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [52] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E279K |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | We confirm the previously described poor prognosis of CML patients with mutations in the BCR-ABL1 kD, since 40.0% of our CML patients who harbored a BCR-ABL1 kD mutation died from CML while receiving TkI treatment. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [27] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.Q252R |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | We identified BCR-ABL kinase domain mutations in 29 of 32 patients whose disease relapsed after an initial response to the tyrosine kinase inhibitor imatinib. Fifteen different amino acid substitutions affecting 13 residues in the kinase domain were found. Mutations fell into two groups-those that alter amino acids that directly contact imatinib and those postulated to prevent BCR-ABL from achieving the inactive conformational state required for imatinib binding. Distinct mutations conferred varying degrees of imatinib resistance. Mutations detected in a subset of patients with stable chronic phase disease correlated with subsequent disease progression. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [27] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.M343T |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | We identified BCR-ABL kinase domain mutations in 29 of 32 patients whose disease relapsed after an initial response to the tyrosine kinase inhibitor imatinib. Fifteen different amino acid substitutions affecting 13 residues in the kinase domain were found. Mutations fell into two groups-those that alter amino acids that directly contact imatinib and those postulated to prevent BCR-ABL from achieving the inactive conformational state required for imatinib binding. Distinct mutations conferred varying degrees of imatinib resistance. Mutations detected in a subset of patients with stable chronic phase disease correlated with subsequent disease progression. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [10] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.K294>RGG |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | BCR-ABL kinase domain mutations were sequentially analyzed in a patient with chronic myeloid leukemia (CML) who exhibited repeated B-lymphoid blast crisis (CML-BC) during treatment with imatinib and dasatinib. We first identified five mutant BCR-ABL clones: Y253H, G250E, F311L, F317L and k294RGG, which was generated by two-nucleotide mutations and six-nucleotide insertion, at the third BC during the imatinib treatment, and retrospectively found that three of them (Y253H, G250E, k294RGG) were already present at the second BC. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.V299L |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Circulating-free DNA assay; Whole exome sequencing assay | |||
Mechanism Description | In patients treated sequentially with dasatinib, nilotinib, or both TkIs after imatinib failure who had developed resistance to second-line treatment, analysis of the individual components of the compound mutations revealed that the identities of component mutations reflected the type of prior drug exposure. Therefore, in all patients treated with dasatinib, at least 1 component of the compound mutations was V299L, F317L, or E255k, all of which have been reported in clinical or in vitro resistance to dasatinib. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F317L |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Circulating-free DNA assay; Whole exome sequencing assay | |||
Mechanism Description | In patients treated sequentially with dasatinib, nilotinib, or both TkIs after imatinib failure who had developed resistance to second-line treatment, analysis of the individual components of the compound mutations revealed that the identities of component mutations reflected the type of prior drug exposure. Therefore, in all patients treated with dasatinib, at least 1 component of the compound mutations was V299L, F317L, or E255k, all of which have been reported in clinical or in vitro resistance to dasatinib. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [11] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E255K |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Circulating-free DNA assay; Whole exome sequencing assay | |||
Mechanism Description | In patients treated sequentially with dasatinib, nilotinib, or both TkIs after imatinib failure who had developed resistance to second-line treatment, analysis of the individual components of the compound mutations revealed that the identities of component mutations reflected the type of prior drug exposure. Therefore, in all patients treated with dasatinib, at least 1 component of the compound mutations was V299L, F317L, or E255k, all of which have been reported in clinical or in vitro resistance to dasatinib. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [53] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.T315N |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
cDNA sequencing assay; Denaturing high-power liquid chromatography assay | |||
Mechanism Description | Our results confirm the high frequency of BCR-ABL kinase domain mutations in patients with secondary resistance to imatinib and exclude mutations of the activation loops of kIT, PDGFRA and PDGFRB as possible causes of resistance in patients without ABL mutations. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [53] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F359A |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
cDNA sequencing assay; Denaturing high-power liquid chromatography assay | |||
Mechanism Description | Our results confirm the high frequency of BCR-ABL kinase domain mutations in patients with secondary resistance to imatinib and exclude mutations of the activation loops of kIT, PDGFRA and PDGFRB as possible causes of resistance in patients without ABL mutations. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [54] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.G398R |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Nested reverse transcriptase polymerase chain reaction assay; Direct sequencing assay | |||
Experiment for Drug Resistance |
Progression-free survival assay; Overall survival assay | |||
Mechanism Description | Two patients had p.E355G mutation in the catalytic domain, and the third patient had p.G398R in the activation loop that is reported here for the first time. Mutation status had no impact on the overall survival and progression-free survival. p.E355G mutation was correlated with shorter survival (P=0.047) in resistant patients. We conclude that BCR- ABL1 mutations are associated with the clinical resistance, but may not be considered the only cause of resistance to imatinib. Mutational analysis may identify resistant patients at risk of disease progression. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [27], [28], [29] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.Y253H |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Allele-specific (AS)-RT-PCR assay | |||
Mechanism Description | We herein describe the development of a rapid allele-specific (AS)-RT-PCR assay to identify the T315I mutation, which confers full resistance to all available tyrosine-kinase inhibitors (TkI). | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [26], [27], [28] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.T315I |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay; Allele-specific (AS)-RT-PCR assay | |||
Mechanism Description | We herein describe the development of a rapid allele-specific (AS)-RT-PCR assay to identify the T315I mutation, which confers full resistance to all available tyrosine-kinase inhibitors (TkI). | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [27], [28], [29] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.Y253F |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
cDNA sequencing assay; Standard dideoxy chain-termination DNA sequencing assay | |||
Experiment for Drug Resistance |
Event-free survival assay; Overall survival assay | |||
Mechanism Description | Mutation scoring can predict outcome in CML-chronic phase with imatinib failure treated with second-generation TkIs and can help in therapy selection. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [27], [28], [29] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.Q252H |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
cDNA sequencing assay; Standard dideoxy chain-termination DNA sequencing assay | |||
Experiment for Drug Resistance |
Event-free survival assay; Overall survival assay | |||
Mechanism Description | Mutation scoring can predict outcome in CML-chronic phase with imatinib failure treated with second-generation TkIs and can help in therapy selection. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [13], [31], [36] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.M388L |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
cDNA sequencing assay; Standard dideoxy chain-termination DNA sequencing assay | |||
Experiment for Drug Resistance |
Event-free survival assay; Overall survival assay | |||
Mechanism Description | Mutation scoring can predict outcome in CML-chronic phase with imatinib failure treated with second-generation TkIs and can help in therapy selection. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [26], [27], [28] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.M351T |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
cDNA sequencing assay; Standard dideoxy chain-termination DNA sequencing assay | |||
Experiment for Drug Resistance |
Event-free survival assay; Overall survival assay | |||
Mechanism Description | Mutation scoring can predict outcome in CML-chronic phase with imatinib failure treated with second-generation TkIs and can help in therapy selection. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [27], [28], [29] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.M244V |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
cDNA sequencing assay; Standard dideoxy chain-termination DNA sequencing assay | |||
Experiment for Drug Resistance |
Event-free survival assay; Overall survival assay | |||
Mechanism Description | Mutation scoring can predict outcome in CML-chronic phase with imatinib failure treated with second-generation TkIs and can help in therapy selection. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [27], [28], [29] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.L387M |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
cDNA sequencing assay; Standard dideoxy chain-termination DNA sequencing assay | |||
Experiment for Drug Resistance |
Event-free survival assay; Overall survival assay | |||
Mechanism Description | Mutation scoring can predict outcome in CML-chronic phase with imatinib failure treated with second-generation TkIs and can help in therapy selection. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [27], [28], [29] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.H396R |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
cDNA sequencing assay; Standard dideoxy chain-termination DNA sequencing assay | |||
Experiment for Drug Resistance |
Event-free survival assay; Overall survival assay | |||
Mechanism Description | Mutation scoring can predict outcome in CML-chronic phase with imatinib failure treated with second-generation TkIs and can help in therapy selection. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [31] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E459Q |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
cDNA sequencing assay; Standard dideoxy chain-termination DNA sequencing assay | |||
Experiment for Drug Resistance |
Event-free survival assay; Overall survival assay | |||
Mechanism Description | Mutation scoring can predict outcome in CML-chronic phase with imatinib failure treated with second-generation TkIs and can help in therapy selection. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [31], [38] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E355A |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
cDNA sequencing assay; Standard dideoxy chain-termination DNA sequencing assay | |||
Experiment for Drug Resistance |
Event-free survival assay; Overall survival assay | |||
Mechanism Description | Mutation scoring can predict outcome in CML-chronic phase with imatinib failure treated with second-generation TkIs and can help in therapy selection. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [28], [29], [53] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E255V |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
cDNA sequencing assay; Standard dideoxy chain-termination DNA sequencing assay | |||
Experiment for Drug Resistance |
Event-free survival assay; Overall survival assay | |||
Mechanism Description | Mutation scoring can predict outcome in CML-chronic phase with imatinib failure treated with second-generation TkIs and can help in therapy selection. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [31], [39], [48] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.D276G |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
cDNA sequencing assay; Standard dideoxy chain-termination DNA sequencing assay | |||
Experiment for Drug Resistance |
Event-free survival assay; Overall survival assay | |||
Mechanism Description | Mutation scoring can predict outcome in CML-chronic phase with imatinib failure treated with second-generation TkIs and can help in therapy selection. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [31], [41] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.A433T |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Peripheral blood | Blood | Homo sapiens (Human) | N.A. |
Bone marrow | Blood | Homo sapiens (Human) | N.A. | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
cDNA sequencing assay; Standard dideoxy chain-termination DNA sequencing assay | |||
Experiment for Drug Resistance |
Event-free survival assay; Overall survival assay | |||
Mechanism Description | Mutation scoring can predict outcome in CML-chronic phase with imatinib failure treated with second-generation TkIs and can help in therapy selection. | |||
|
||||
Key Molecule: hsa-mir-328 | [55] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | KG-1 cells | Bone marrow | Homo sapiens (Human) | CVCL_0374 |
K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | LncRNA MALAT1 promotes cell proliferation and imatinib resistance by suppressing miR-328 in chronic myelogenous leukemia. | |||
Key Molecule: Metastasis associated lung adenocarcinoma transcript 1 (MALAT1) | [55] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | KG-1 cells | Bone marrow | Homo sapiens (Human) | CVCL_0374 |
K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | LncRNA MALAT1 promotes cell proliferation and imatinib resistance by suppressing miR-328 in chronic myelogenous leukemia. | |||
Key Molecule: hsa_circ_BA9.3 | [14] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Ku812 cells | Bone marrow | Homo sapiens (Human) | CVCL_0379 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCk reagent assay; Flow cytometry assay | |||
Mechanism Description | CircBA9.3 promoted cell proliferation and reduced the sensitivity of leukaemic cells to TkIs through up-regulation of the ABL1 and BCR-ABL1 protein expression levels. | |||
Key Molecule: hsa-miR-205-5p | [56] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | ABCC2 was a downstream target of miR205-5p, overexpression of miR205-5p suppressed the expression of ABCC2 in k562-R cells. SNHG5 promotes imatinib resistance through upregulating ABCC2. SNHG5 promotes imatinib resistance in CML via acting as a competing endogenous RNA against miR205-5p. | |||
Key Molecule: Small nucleolar RNA host gene 5 (SNHG5) | [56] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | SNHG5 promotes imatinib resistance through upregulating ABCC2 and promotes imatinib resistance in CML via acting as a competing endogenous RNA against miR205-5p. | |||
Key Molecule: hsa-miR-29a-3p | [15] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Lin-CD34+CD38- cells | Bone | Homo sapiens (Human) | N.A. |
Lin-CD34-CD38- CML cells | Bone | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Annexin V assay | |||
Mechanism Description | The up-regulation of miR29a-3p observed in CML LSCs led to the down-regulation of its target TET2 and conferred TkI-resistance to CML LSCs in vitro. | |||
Key Molecule: hsa-miR-494-3p | [15] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Lin-CD34+CD38- cells | Bone | Homo sapiens (Human) | N.A. |
Lin-CD34-CD38- CML cells | Bone | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Annexin V assay | |||
Mechanism Description | miR494-3p down-regulation in CML LSCs, leading to c-MYC up-regulation, was able to decrease TkI-induced apoptosis. | |||
Key Molecule: hsa-miR-660-5p | [15] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Lin-CD34+CD38- cells | Bone | Homo sapiens (Human) | N.A. |
Lin-CD34-CD38- CML cells | Bone | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Annexin V assay | |||
Mechanism Description | The up-regulation of miR660-5p observed in CML LSCs led to the down-regulation of its target EPAS1 and conferred TkI-resistance to CML LSCs in vitro. | |||
Key Molecule: hsa-let-7i | [57] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTS assay; Flow cytometric analysis; CFU assay | |||
Mechanism Description | miR224 and let-7i regulate the proliferation and chemosensitivity of CML cells probably via targeting ST3GAL IV. | |||
Key Molecule: hsa-mir-224 | [57] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTS assay; Flow cytometric analysis; CFU assay | |||
Mechanism Description | miR224 and let-7i regulate the proliferation and chemosensitivity of CML cells probably via targeting ST3GAL IV. | |||
Key Molecule: Hepatocellular carcinoma up-regulated long non-coding RNA (HULC) | [58] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell invasion | Activation | hsa05200 | |
Cell proliferation | Activation | hsa05200 | ||
PI3K/AKT signaling pathway | Activation | hsa04151 | ||
In Vitro Model | KG-1 cells | Bone marrow | Homo sapiens (Human) | CVCL_0374 |
THP-1 cells | Blood | Homo sapiens (Human) | CVCL_0006 | |
K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 | |
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | Long noncoding RNA HULC promotes cell proliferation by regulating PI3k/AkT signaling pathway in chronic myeloid leukemia. HULC aggrevates CML by regulating PI3k/AkT. Inhibition of HULC enhances imatinib induced CML apoptosis. 3. HULC increased c-Myc and Bcl-2 by sequestering miR200a-3p. | |||
Key Molecule: HOX transcript antisense RNA (HOTAIR) | [59] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | PI3K/AKT signaling pathway | Activation | hsa04151 | |
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
K562-R cells | Pleural effusion | Homo sapiens (Human) | CVCL_5950 | |
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay; Annexin V/propidium iodide staining assay | |||
Mechanism Description | Knockdown of HOTAIR expression downregulated the MRP1 expression levels in the k562-imatinib cells and resulted in higher sensitivity to the imatinib treatment. In addition, the activation of PI3k/Akt was greatly attenuated when HOTAIR was knocked down in k562-imatinib cells. | |||
Key Molecule: hsa-mir-16 | [60] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | LncRNA UCA1 Contributes to Imatinib Resistance by Acting as a ceRNA Against miR16 in Chronic Myeloid Leukemia Cells. UCA1 directly interacts with miR16. | |||
Key Molecule: Urothelial cancer associated 1 (UCA1) | [60] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | UCA1 functions as a ceRNA of MDR1, UCA1 promotes IM resistance of CML cell through regulation of MDR1. Ectopic expression of MDR1 or silence of miR16 partially rescued this suppression induced by UCA1 knockdown. | |||
Key Molecule: hsa-miR-486-5p | [61] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | TF-1 cells | Bone marrow | Homo sapiens (Human) | CVCL_0559 |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | miR-486-5p expression contributes to survival of BCR-ABL-transformed cells after imatinib treatment and that inhibition of miR-486-5p enhances the sensitivity of CML progenitors to imatinib-mediated apoptosis. | |||
Key Molecule: hsa-mir-199b | [62] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | RT-PCR was found to be a more sensitive technique to study miRNA expression in 9q deleted patients where deletions are missed out by FISH. The miRNA expression is important in the 9q deleted patients as miR-199b associated with drug resistance and can be used as a prognostic factor in 9q deleted CML patients. | |||
Key Molecule: hsa-mir-181c | [63] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
RT-qPCR | |||
Experiment for Drug Resistance |
Response evaluation criteria in solid tumors assay | |||
Mechanism Description | Significant down-regulation of miR-181c in imatinib-resistant versus imatinib-responders was confirmed by qRT-PCR. Some miR-181c target genes such as PBX3, HSP90B1, NMT2 and RAD21 have been associated with drug response. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [9] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.D444Y |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | We confirmed the high frequency of SFks involvement in Tyrosine kinase inhibitor-resistant CML (52% of the cases) and even further in progressive disease and blast crises (60% of the cases). The SFks deregulation is also observed in patients harboring BCR-ABL mutations. In T315I and F317L mutated patients, CML-resistance appears to be promoted by SFks kinase protein reactivation once the BCR-ABL mutated clone has decreased on Omacetaxine. | |||
|
||||
Key Molecule: ATP-binding cassette sub-family C2 (ABCC2) | [56] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
Western blot analysis; Luciferase reporter assay | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | ABCC2 was a downstream target of miR205-5p, overexpression of miR205-5p suppressed the expression of ABCC2 in k562-R cells. SNHG5 promotes imatinib resistance through upregulating ABCC2. SNHG5 promotes imatinib resistance in CML via acting as a competing endogenous RNA against miR205-5p. | |||
Key Molecule: Multidrug resistance-associated protein 1 (MRP1) | [59] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | PI3K/AKT signaling pathway | Activation | hsa04151 | |
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
K562-R cells | Pleural effusion | Homo sapiens (Human) | CVCL_5950 | |
Experiment for Molecule Alteration |
qRT-PCR; Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay; Annexin V/propidium iodide staining assay | |||
Mechanism Description | Knockdown of HOTAIR expression downregulated the MRP1 expression levels in the k562-imatinib cells and resulted in higher sensitivity to the imatinib treatment. In addition, the activation of PI3k/Akt was greatly attenuated when HOTAIR was knocked down in k562-imatinib cells. | |||
Key Molecule: Multidrug resistance protein 1 (ABCB1) | [60] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay | |||
Mechanism Description | UCA1 functions as a ceRNA of MDR1, UCA1 promotes IM resistance of CML cell through regulation of MDR1. Ectopic expression of MDR1 or silence of miR16 partially rescued this suppression induced by UCA1 knockdown. | |||
|
||||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [14] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Ku812 cells | Bone marrow | Homo sapiens (Human) | CVCL_0379 | |
Experiment for Molecule Alteration |
Western blot analysis; qRT-PCR | |||
Experiment for Drug Resistance |
CCk reagent assay; Flow cytometry assay | |||
Mechanism Description | CircBA9.3 promoted cell proliferation and reduced the sensitivity of leukaemic cells to TkIs through up-regulation of the ABL1 and BCR-ABL1 protein expression levels. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [14] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Ku812 cells | Bone marrow | Homo sapiens (Human) | CVCL_0379 | |
Experiment for Molecule Alteration |
Western blot analysis; qRT-PCR | |||
Experiment for Drug Resistance |
CCk reagent assay; Flow cytometry assay | |||
Mechanism Description | CircBA9.3 promoted cell proliferation and reduced the sensitivity of leukaemic cells to TkIs through up-regulation of the ABL1 and BCR-ABL1 protein expression levels. | |||
Key Molecule: Myc proto-oncogene protein (MYC) | [15] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Lin-CD34+CD38- cells | Bone | Homo sapiens (Human) | N.A. |
Lin-CD34-CD38- CML cells | Bone | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Annexin V assay | |||
Mechanism Description | miR494-3p down-regulation in CML LSCs, leading to c-MYC up-regulation, was able to decrease TkI-induced apoptosis. | |||
Key Molecule: Hypoxia-inducible factor 2-alpha (EPAS1) | [15] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Lin-CD34+CD38- cells | Bone | Homo sapiens (Human) | N.A. |
Lin-CD34-CD38- CML cells | Bone | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Annexin V assay | |||
Mechanism Description | The up-regulation of miR660-5p observed in CML LSCs led to the down-regulation of its target EPAS1 and conferred TkI-resistance to CML LSCs in vitro. | |||
Key Molecule: Methylcytosine dioxygenase TET2 (TET2) | [15] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Lin-CD34+CD38- cells | Bone | Homo sapiens (Human) | N.A. |
Lin-CD34-CD38- CML cells | Bone | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Annexin V assay | |||
Mechanism Description | The up-regulation of miR29a-3p observed in CML LSCs led to the down-regulation of its target TET2 and conferred TkI-resistance to CML LSCs in vitro. | |||
Key Molecule: Sialyltransferase 4C (SIAT4C) | [57] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTS assay; Flow cytometric analysis; CFU assay | |||
Mechanism Description | miR224 and let-7i regulate the proliferation and chemosensitivity of CML cells probably via targeting ST3GAL IV. | |||
Key Molecule: Myc proto-oncogene protein (MYC) | [58] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
PI3K/AKT signaling pathway | Activation | hsa04151 | ||
In Vitro Model | KG-1 cells | Bone marrow | Homo sapiens (Human) | CVCL_0374 |
THP-1 cells | Blood | Homo sapiens (Human) | CVCL_0006 | |
K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometry assay | |||
Mechanism Description | Long noncoding RNA HULC promotes cell proliferation by regulating PI3k/AkT signaling pathway in chronic myeloid leukemia. HULC aggrevates CML by regulating PI3k/AkT. Inhibition of HULC enhances imatinib induced CML apoptosis. 3. HULC increased c-Myc and Bcl-2 by sequestering miR200a-3p. | |||
Key Molecule: NUP98-DDX10 fusion protein type 1 (NUP98-DDX10 ) | [64] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Structural mutation | Structural variation |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
RT-PCR analysis | |||
Experiment for Drug Resistance |
Western blot analysis with anti-CrkL antibody assay | |||
Mechanism Description | Collectively, these observations raise the possibility that NUP98/DDX10 might have played a role not only in disease progression but also in the acquisition of resistance to imatinib. | |||
Key Molecule: GTPase Nras (NRAS) | [4], [5] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.G12V |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | JAKT2/STAT signaling pathway | Activation | hsa04030 | |
RAF/KRAS/MEK signaling pathway | Activation | hsa04010 | ||
In Vitro Model | HL60 cells | Peripheral blood | Homo sapiens (Human) | CVCL_0002 |
U937 cells | Blood | Homo sapiens (Human) | CVCL_0007 | |
K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 | |
KCL-22 cells | Bone marrow | Homo sapiens (Human) | CVCL_2091 | |
Sup-B15 cells | Bone marrow | Homo sapiens (Human) | CVCL_0103 | |
HEL cells | Blood | Homo sapiens (Human) | CVCL_0001 | |
HMC-1.2 cells | Blood | Homo sapiens (Human) | CVCL_H205 | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Next-generation sequencing assay; Sanger Sequencing assay | |||
Mechanism Description | This mutation is well known for its effects on proliferation and its association with AML and MPN, suggesting that this variant might have been involved in the TkI resistance of this patient. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [36] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.R328M |
||
Resistant Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
DNA sequencing assay | |||
Experiment for Drug Resistance |
Progression-free survival assay | |||
Mechanism Description | We conclude that the currently recommended 10-fold threshold to trigger mutation screening is insensitive and not universally applicable. kinase domain mutations predict a shorter progression-free survival. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: hsa-mir-7 | [65] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | BCR-ABL/PI3K/AKT signaling pathway | Inhibition | hsa05220 | |
Cell apoptosis | Activation | hsa04210 | ||
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometric analysis | |||
Mechanism Description | miR7 inhibits cell proliferation and increases cell apoptosis in k562 cells and downregulates BCR-ABL/PI3k/AkT signaling in k562 cells, thus sensitizing k562 cells to imatinib. | |||
Key Molecule: Maternally expressed 3 (MEG3) | [66] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
RT-qPCR | |||
Experiment for Drug Resistance |
CCK8 assay; Annexin V-FITC/PI Apoptosis Detection assay | |||
Mechanism Description | LncRNA MEG3 Regulates Imatinib Resistance in Chronic Myeloid Leukemia via Suppressing microRNA-21. MEG3 and miR21 were negatively correlated in CML patients, miR21 mimics reversed the phenotype of MEG3-overexpression in imatinib-resistant k562 cells. | |||
Key Molecule: Nuclear paraspeckle assembly transcript 1 (NEAT1) | [67] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | HL60 cells | Peripheral blood | Homo sapiens (Human) | CVCL_0002 |
Jurkat cells | Pleural effusion | Homo sapiens (Human) | CVCL_0065 | |
K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 | |
MOLT4 cells | Bone marrow | Homo sapiens (Human) | CVCL_0013 | |
NB4 cells | Bone marrow | Homo sapiens (Human) | CVCL_0005 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | The c-Myc-regulated LncRNA NEAT1 and paraspeckles modulate imatinib-induced apoptosis in CML cells. | |||
Key Molecule: hsa-miR-199a-5p | [68] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell viability | Inhibition | hsa05200 | ||
Wnt2-mediated Beta-catenin signaling pathway | Inhibition | hsa04310 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Ku812 cells | Bone marrow | Homo sapiens (Human) | CVCL_0379 | |
Experiment for Molecule Alteration |
RT-qPCR | |||
Experiment for Drug Resistance |
MTT assay; Flow cytometry assay | |||
Mechanism Description | microRNA-199a/b-5p enhance imatinib efficacy via repressing WNT2 signaling-mediated protective autophagy in imatinib-resistant chronic myeloid leukemia cells. | |||
Key Molecule: hsa-miR-199b-5p | [68] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell viability | Inhibition | hsa05200 | ||
Wnt2-mediated Beta-catenin signaling pathway | Inhibition | hsa04310 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Ku812 cells | Bone marrow | Homo sapiens (Human) | CVCL_0379 | |
Experiment for Molecule Alteration |
RT-qPCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | microRNA-199a/b-5p enhance imatinib efficacy via repressing WNT2 signaling-mediated protective autophagy in imatinib-resistant chronic myeloid leukemia cells. | |||
Key Molecule: hsa-mir-202 | [69] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell viability | Activation | hsa05200 | |
In Vitro Model | HL60 cells | Peripheral blood | Homo sapiens (Human) | CVCL_0002 |
K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 | |
Ku812 cells | Bone marrow | Homo sapiens (Human) | CVCL_0379 | |
KCL-22 cells | Bone marrow | Homo sapiens (Human) | CVCL_2091 | |
EM2 cells | Bone | Homo sapiens (Human) | CVCL_1196 | |
EM3 cells | Bone | Homo sapiens (Human) | CVCL_2033 | |
LAMA 84 cells | Bone | Homo sapiens (Human) | CVCL_0388 | |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay; BrdU assay; Caspase-3 assay | |||
Mechanism Description | Overexpression of miR-202 sensitized imatinib resistant CML through the miR-202-mediated glycolysis inhibition by targetting Hk2. | |||
Key Molecule: hsa-mir-1301 | [70] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Ku812 cells | Bone marrow | Homo sapiens (Human) | CVCL_0379 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | microRNA-1301-mediated RanGAP1 downregulation induces BCR-ABL nuclear entrapment to enhance imatinib efficacy in chronic myeloid leukemia cells. | |||
Key Molecule: hsa-mir-130a | [71] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
p53 signaling pathway | Regulation | hsa04115 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | BCL-2, MCL-1 and XIAP were the target genes of miR-130a. BCL-2, MCL-1, TCL-1 and XIAP protein levels were significantly higher in patients with drug-sensitive CML cells. Transfected miR-130a mimics significantly decreased the protein expression of BCL-1, MCL-1 and XIAP. Transfected miR-130a significantly increased the CML sensitivity to Gleevec. | |||
Key Molecule: hsa-mir-30e | [72] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
JAKT/STAT/PI3K/AKT signaling pathway | Inhibition | hsa04630 | ||
In Vitro Model | THP-1 cells | Blood | Homo sapiens (Human) | CVCL_0006 |
HL60 cells | Peripheral blood | Homo sapiens (Human) | CVCL_0002 | |
K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 | |
HEK293 cells | Kidney | Homo sapiens (Human) | CVCL_0045 | |
Meg-01 cells | Blood | Homo sapiens (Human) | CVCL_0425 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
WST-1 assay | |||
Mechanism Description | Luciferase assay verified that miR-30e directly targets ABL. Enforced expression of miR-30e in k562 cells suppressed proliferation and induced apoptosis of these cells and sensitized them to imatinib treatment. These findings strongly suggest that miR-30e acts as a tumor suppressor by downregulating BCR-ABL expression. | |||
Key Molecule: hsa-mir-30a | [73], [74] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Intrinsic apoptotic signaling pathway | Activation | hsa04210 | ||
Mitochondrial signaling pathway | Activation | hsa04217 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
qPCR | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | miR-30a mimic or knockdown of autophagy genes (ATGs) such as Beclin 1 and ATG5 by short hairpin RNA enhances imatinib-induced cytotoxicity and promotes mitochondria-dependent intrinsic apoptosis. In contrast, knockdown of miR-30a by antagomiR-30a increases the expression of Beclin 1 and ATG5, and inhibits imatinib-induced cytotoxicity. And MIR30A mimics, as well as knockdown of BECN1 and ATG5, increases intrinsic apoptotic pathways. | |||
Key Molecule: hsa-mir-203 | [75] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell proliferation | Inhibition | hsa05200 | |
In Vitro Model | BaF3-BCR/ABLT315I cells | Bone marrow | Homo sapiens (Human) | CVCL_UE64 |
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Interference BCR/ABL expression with miR-203 restored the sensitivity to imatinib in cells expressing the imatinib-resistant BCR/ABL kinase domain mutant T315I. | |||
Key Molecule: hsa-mir-144 | [76] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | c-Myc expression was upregulated in the imatinib resistant k562R cells, which in turn increased the expression of miR-144/451, restoration of miR-144/451 or knockdown of Myc could sensitize the imatinib resistant cells to apoptosis. Myc, miR-144/451 form a regulatory pathway and contribute to the imatinib resistance. | |||
Key Molecule: hsa-mir-451 | [76] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | c-Myc expression was upregulated in the imatinib resistant k562R cells, which in turn increased the expression of miR-144/451, restoration of miR-144/451 or knockdown of Myc could sensitize the imatinib resistant cells to apoptosis. Myc, miR-144/451 form a regulatory pathway and contribute to the imatinib resistance. | |||
Key Molecule: hsa-mir-21 | [66] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
RT-qPCR | |||
Experiment for Drug Resistance |
CCK8 assay; Annexin V-FITC/PI Apoptosis Detection assay | |||
Mechanism Description | LncRNA MEG3 regulates imatinib resistance in chronic myeloid leukemia via suppressing microRNA-21. MEG3 and miR21 were negatively correlated in CML patients, miR21 mimics reversed the phenotype of MEG3-overexpression in imatinib-resistant k562 cells. | |||
|
||||
Key Molecule: Multidrug resistance-associated protein 1 (MRP1) | [66] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay; Annexin V-FITC/PI Apoptosis Detection assay | |||
Mechanism Description | Overexpression of MEG3 in imatinib-resistant k562 cells markedly decreased cell proliferation, increased cell apoptosis, reversed imatinib resistance, and reduced the expression of MRP1, MDR1, and ABCG2. | |||
Key Molecule: ATP-binding cassette sub-family G2 (ABCG2) | [66] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay; Annexin V-FITC/PI Apoptosis Detection assay | |||
Mechanism Description | Overexpression of MEG3 in imatinib-resistant k562 cells markedly decreased cell proliferation, increased cell apoptosis, reversed imatinib resistance, and reduced the expression of MRP1, MDR1, and ABCG2. | |||
Key Molecule: Multidrug resistance protein 1 (ABCB1) | [66] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
CCK8 assay; Annexin V-FITC/PI Apoptosis Detection assay | |||
Mechanism Description | Overexpression of MEG3 in imatinib-resistant k562 cells markedly decreased cell proliferation, increased cell apoptosis, reversed imatinib resistance, and reduced the expression of MRP1, MDR1, and ABCG2. | |||
|
||||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [65] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | BCR-ABL/PI3K/AKT signaling pathway | Inhibition | hsa05220 | |
Cell apoptosis | Activation | hsa04210 | ||
Cell proliferation | Inhibition | hsa05200 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
Western blot analysis; Dual luciferase reporter assay | |||
Experiment for Drug Resistance |
CCK8 assay; Flow cytometric analysis | |||
Mechanism Description | miR7 inhibits cell proliferation and increases cell apoptosis in k562 cells and downregulates BCR-ABL/PI3k/AkT signaling in k562 cells, thus sensitizing k562 cells to imatinib. | |||
Key Molecule: Int-1-related protein (WNT2) | [68] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell colony | Inhibition | hsa05200 | ||
Cell viability | Inhibition | hsa05200 | ||
Wnt2-mediated Beta-catenin signaling pathway | Inhibition | hsa04310 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Ku812 cells | Bone marrow | Homo sapiens (Human) | CVCL_0379 | |
Experiment for Molecule Alteration |
Western blot analysis; RIP assay; Luciferase reporter assay | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | microRNA-199a/b-5p enhance imatinib efficacy via repressing WNT2 signaling-mediated protective autophagy in imatinib-resistant chronic myeloid leukemia cells. | |||
Key Molecule: Hexokinase-2 (HK2) | [69] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell viability | Activation | hsa05200 | |
In Vitro Model | HL60 cells | Peripheral blood | Homo sapiens (Human) | CVCL_0002 |
K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 | |
Ku812 cells | Bone marrow | Homo sapiens (Human) | CVCL_0379 | |
KCL-22 cells | Bone marrow | Homo sapiens (Human) | CVCL_2091 | |
EM2 cells | Bone | Homo sapiens (Human) | CVCL_1196 | |
EM3 cells | Bone | Homo sapiens (Human) | CVCL_2033 | |
LAMA 84 cells | Bone | Homo sapiens (Human) | CVCL_0388 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay; BrdU assay; Caspase-3 assay | |||
Mechanism Description | Overexpression of miR-202 sensitized imatinib resistant CML through the miR-202-mediated glycolysis inhibition by targetting Hk2. | |||
Key Molecule: Ran GTPase-activating protein 1 (RANGAP1) | [70] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Ku812 cells | Bone marrow | Homo sapiens (Human) | CVCL_0379 | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | microRNA-1301-mediated RanGAP1 downregulation induces BCR-ABL nuclear entrapment to enhance imatinib efficacy in chronic myeloid leukemia cells. | |||
Key Molecule: Apoptosis regulator Bcl-2 (BCL2) | [71] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
p53 signaling pathway | Regulation | hsa04115 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | BCL-2, MCL-1 and XIAP were the target genes of miR-130a. BCL-2, MCL-1, TCL-1 and XIAP protein levels were significantly higher in patients with drug-sensitive CML cells. Transfected miR-130a mimics significantly decreased the protein expression of BCL-1, MCL-1 and XIAP. Transfected miR-130a significantly increased the CML sensitivity to Gleevec. | |||
Key Molecule: Induced myeloid leukemia cell differentiation protein Mcl-1 (MCL1) | [71] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
p53 signaling pathway | Regulation | hsa04115 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | BCL-2, MCL-1 and XIAP were the target genes of miR-130a. BCL-2, MCL-1, TCL-1 and XIAP protein levels were significantly higher in patients with drug-sensitive CML cells. Transfected miR-130a mimics significantly decreased the protein expression of BCL-1, MCL-1 and XIAP. Transfected miR-130a significantly increased the CML sensitivity to Gleevec. | |||
Key Molecule: E3 ubiquitin-protein ligase XIAP (XIAP) | [71] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
p53 signaling pathway | Regulation | hsa04115 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | BCL-2, MCL-1 and XIAP were the target genes of miR-130a. BCL-2, MCL-1, TCL-1 and XIAP protein levels were significantly higher in patients with drug-sensitive CML cells. Transfected miR-130a mimics significantly decreased the protein expression of BCL-1, MCL-1 and XIAP. Transfected miR-130a significantly increased the CML sensitivity to Gleevec. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [72] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Cell proliferation | Inhibition | hsa05200 | ||
JAKT/STAT/PI3K/AKT signaling pathway | Inhibition | hsa04630 | ||
In Vitro Model | THP-1 cells | Blood | Homo sapiens (Human) | CVCL_0006 |
HL60 cells | Peripheral blood | Homo sapiens (Human) | CVCL_0002 | |
K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 | |
HEK293 cells | Kidney | Homo sapiens (Human) | CVCL_0045 | |
Meg-01 cells | Blood | Homo sapiens (Human) | CVCL_0425 | |
In Vivo Model | Nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
WST-1 assay | |||
Mechanism Description | Luciferase assay verified that miR-30e directly targets ABL. Enforced expression of miR-30e in k562 cells suppressed proliferation and induced apoptosis of these cells and sensitized them to imatinib treatment. These findings strongly suggest that miR-30e acts as a tumor suppressor by downregulating BCR-ABL expression. | |||
Key Molecule: Autophagy protein 5 (ATG5) | [73] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Mitochondrial signaling pathway | Activation | hsa04217 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | miR-30a mimic or knockdown of autophagy genes (ATGs) such as Beclin 1 and ATG5 by short hairpin RNA enhances imatinib-induced cytotoxicity and promotes mitochondria-dependent intrinsic apoptosis. In contrast, knockdown of miR-30a by antagomiR-30a increases the expression of Beclin 1 and ATG5, and inhibits imatinib-induced cytotoxicity. | |||
Key Molecule: Beclin-1 (BECN1) | [73] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Imatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
Mitochondrial signaling pathway | Activation | hsa04217 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | miR-30a mimic or knockdown of autophagy genes (ATGs) such as Beclin 1 and ATG5 by short hairpin RNA enhances imatinib-induced cytotoxicity and promotes mitochondria-dependent intrinsic apoptosis. In contrast, knockdown of miR-30a by antagomiR-30a increases the expression of Beclin 1 and ATG5, and inhibits imatinib-induced cytotoxicity. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: ATP-binding cassette sub-family G2 (ABCG2) | [77] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Mitoxantrone | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | K562/BCRP cells | Blood | Homo sapiens (Human) | CVCL_0004 |
K562/MDR cells | Blood | Homo sapiens (Human) | CVCL_0004 | |
Experiment for Drug Resistance |
Growth inhibition assay | |||
Mechanism Description | Some flavonoids have BCRP-inhibitory activity. 3',4',7-trimethoxyflavone showed the strongest anti-BCRP activity with RI50 values of 0.012 uM for SN-38 and 0.044 uM for mitoxantrone. 3',4',7-trimethoxyflavone and acacetin, showed only low anti-P-gp activity, with the remainder displaying no suppressive effects against P-gp. None of the flavonoids that we tested inhibite. |
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [7] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.M351T |
||
Resistant Drug | Nilotinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Progression-free survival assay; Overall survival assay | |||
Mechanism Description | After 13 months of therapy a progression of disease to accelerated phase was detected and a second mutational screening analysis performed at that time revealed the absence of M244 V and the appearance of M351T mutation. After 14 months of therapy, a third mutational analysis was performed which revealed the disappearance of M351T mutation and the acquisition of a new F317L mutation. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [41] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F359C |
||
Resistant Drug | Nilotinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Next generation sequencing assay | |||
Experiment for Drug Resistance |
Event-free survival assay; Overall survival assay | |||
Mechanism Description | HSCT is an important salvage option for TkI-resistant patients with or without BCR-ABL1 mutations. Patients with mutations were more likely to develop advanced disease and had worse outcomes after HSCT. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [41] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E255V |
||
Resistant Drug | Nilotinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Next generation sequencing assay | |||
Experiment for Drug Resistance |
Event-free survival assay; Overall survival assay | |||
Mechanism Description | HSCT is an important salvage option for TkI-resistant patients with or without BCR-ABL1 mutations. Patients with mutations were more likely to develop advanced disease and had worse outcomes after HSCT. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [41] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E255K |
||
Resistant Drug | Nilotinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Next generation sequencing assay | |||
Experiment for Drug Resistance |
Event-free survival assay; Overall survival assay | |||
Mechanism Description | HSCT is an important salvage option for TkI-resistant patients with or without BCR-ABL1 mutations. Patients with mutations were more likely to develop advanced disease and had worse outcomes after HSCT. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [13], [78] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.T315I |
||
Resistant Drug | Nilotinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | Our finding reflects the natural development of a T315I mutation within the hematopoietic system of the reported patient and indicates the importance of BCR-ABL1 mutation monitoring in more primitive cell populations. Considering the natural history of T315I development in this reported CML case, we hypothesize that BCR-ABL1 kD mutations may be pre-concentrated in more primitive CML cells, which subsequently expand into the PB. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [8] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F359I |
||
Resistant Drug | Nilotinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Mechanism Description | The most common mechanism of acquired resistance in CML in imatinib era is the acquisition of BCR-ABL kinase domain mutations with decreased sensitivity to the drug. Our findings demonstrate the potential hazards of sequential kinase inhibitor therapy and suggest a role for a combination of ABL kinase inhibitors, perhaps including drugs with different mechanisms of action, to prevent the outgrowth of cells harboring drug-resistant BCR-ABL mutations. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [13] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.L387M |
||
Resistant Drug | Nilotinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
RNA sequencing assay | |||
Mechanism Description | The presence of BCR-ABL oncogene mutations in patients with chronic myeloid leukemia (CML) may be responsible for the failure of tyrosine kinase inhibitor (TkI) treatment. In addition to 9 point mutations (G250E / F317L, F359V, L387M, Y253H, M388L, M244V, T315I, D276G), 35 bp insertion between exons 8 and 9 and deletion exon 7 were detected. Our results demonstrate that direct sequencing is suitable for routine clinical monitoring patients with CML and may be useful for optimizing therapy. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [2], [8], [13] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.G250E |
||
Resistant Drug | Nilotinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
RNA sequencing assay | |||
Mechanism Description | The presence of BCR-ABL oncogene mutations in patients with chronic myeloid leukemia (CML) may be responsible for the failure of tyrosine kinase inhibitor (TkI) treatment. In addition to 9 point mutations (G250E / F317L, F359V, L387M, Y253H, M388L, M244V, T315I, D276G), 35 bp insertion between exons 8 and 9 and deletion exon 7 were detected. Our results demonstrate that direct sequencing is suitable for routine clinical monitoring patients with CML and may be useful for optimizing therapy. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [13] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F359V |
||
Resistant Drug | Nilotinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
RNA sequencing assay | |||
Mechanism Description | The presence of BCR-ABL oncogene mutations in patients with chronic myeloid leukemia (CML) may be responsible for the failure of tyrosine kinase inhibitor (TkI) treatment. In addition to 9 point mutations (G250E / F317L, F359V, L387M, Y253H, M388L, M244V, T315I, D276G), 35 bp insertion between exons 8 and 9 and deletion exon 7 were detected. Our results demonstrate that direct sequencing is suitable for routine clinical monitoring patients with CML and may be useful for optimizing therapy. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [13] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F317L |
||
Resistant Drug | Nilotinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
RNA sequencing assay | |||
Mechanism Description | The presence of BCR-ABL oncogene mutations in patients with chronic myeloid leukemia (CML) may be responsible for the failure of tyrosine kinase inhibitor (TkI) treatment. In addition to 9 point mutations (G250E / F317L, F359V, L387M, Y253H, M388L, M244V, T315I, D276G), 35 bp insertion between exons 8 and 9 and deletion exon 7 were detected. Our results demonstrate that direct sequencing is suitable for routine clinical monitoring patients with CML and may be useful for optimizing therapy. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [13] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.D276G |
||
Resistant Drug | Nilotinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
RNA sequencing assay | |||
Mechanism Description | The presence of BCR-ABL oncogene mutations in patients with chronic myeloid leukemia (CML) may be responsible for the failure of tyrosine kinase inhibitor (TkI) treatment. In addition to 9 point mutations (G250E / F317L, F359V, L387M, Y253H, M388L, M244V, T315I, D276G), 35 bp insertion between exons 8 and 9 and deletion exon 7 were detected. Our results demonstrate that direct sequencing is suitable for routine clinical monitoring patients with CML and may be useful for optimizing therapy. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [13], [52] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.Y253H |
||
Resistant Drug | Nilotinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | We confirm the previously described poor prognosis of CML patients with mutations in the BCR-ABL1 kD, since 40.0% of our CML patients who harbored a BCR-ABL1 kD mutation died from CML while receiving TkI treatment. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [52] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.H396R |
||
Resistant Drug | Nilotinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | We confirm the previously described poor prognosis of CML patients with mutations in the BCR-ABL1 kD, since 40.0% of our CML patients who harbored a BCR-ABL1 kD mutation died from CML while receiving TkI treatment. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [3] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.T315V |
||
Resistant Drug | Nilotinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Experiment for Drug Resistance |
Tritiated thymidine incorporation assay | |||
Mechanism Description | Mutations may impair TkI activity by directly or indirectly impairing the drug binding to the protein. We report the discovery of three new BCR/ABL mutations, L248R, T315V, and F317R identified in two patients with CML (L248R and T315V) and in one patient with Ph+ acute lymphoblastic leukemia (ALL) (F317R). | |||
|
||||
Key Molecule: hsa_circ_BA9.3 | [14] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Nilotinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Ku812 cells | Bone marrow | Homo sapiens (Human) | CVCL_0379 | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
CCk reagent assay; Flow cytometry assay | |||
Mechanism Description | CircBA9.3 promoted cell proliferation and reduced the sensitivity of leukaemic cells to TkIs through up-regulation of the ABL1 and BCR-ABL1 protein expression levels. | |||
|
||||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [14] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Nilotinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Ku812 cells | Bone marrow | Homo sapiens (Human) | CVCL_0379 | |
Experiment for Molecule Alteration |
Western blot analysis; qRT-PCR | |||
Experiment for Drug Resistance |
CCk reagent assay; Flow cytometry assay | |||
Mechanism Description | CircBA9.3 promoted cell proliferation and reduced the sensitivity of leukaemic cells to TkIs through up-regulation of the ABL1 and BCR-ABL1 protein expression levels. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [14] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Nilotinib | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
Cell proliferation | Activation | hsa05200 | ||
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Ku812 cells | Bone marrow | Homo sapiens (Human) | CVCL_0379 | |
Experiment for Molecule Alteration |
Western blot analysis; qRT-PCR | |||
Experiment for Drug Resistance |
CCk reagent assay; Flow cytometry assay | |||
Mechanism Description | CircBA9.3 promoted cell proliferation and reduced the sensitivity of leukaemic cells to TkIs through up-regulation of the ABL1 and BCR-ABL1 protein expression levels. | |||
Key Molecule: GTPase Nras (NRAS) | [4], [5] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.G12V |
||
Resistant Drug | Nilotinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | JAKT2/STAT signaling pathway | Activation | hsa04030 | |
RAF/KRAS/MEK signaling pathway | Activation | hsa04010 | ||
In Vitro Model | HL60 cells | Peripheral blood | Homo sapiens (Human) | CVCL_0002 |
U937 cells | Blood | Homo sapiens (Human) | CVCL_0007 | |
K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 | |
KCL-22 cells | Bone marrow | Homo sapiens (Human) | CVCL_2091 | |
Sup-B15 cells | Bone marrow | Homo sapiens (Human) | CVCL_0103 | |
HEL cells | Blood | Homo sapiens (Human) | CVCL_0001 | |
HMC-1.2 cells | Blood | Homo sapiens (Human) | CVCL_H205 | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Next-generation sequencing assay; Sanger Sequencing assay | |||
Mechanism Description | This mutation is well known for its effects on proliferation and its association with AML and MPN, suggesting that this variant might have been involved in the TkI resistance of this patient. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: hsa-mir-217 | [17] | |||
Sensitive Disease | Chronic myelogenous Ph(+) leukemia [ICD-11: 2A20.1] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Nilotinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Bcr/Abl-expressing k562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
K562DR cells | Blood | Homo sapiens (Human) | CVCL_4V59 | |
K562NR cells | Blood | Homo sapiens (Human) | CVCL_4V63 | |
In Vivo Model | BALB/c nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
RT-PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Forced expression of miR-217 inhibited expression of DNMT3A through a miR-217-binding site within the 3'-untranslated region of DNMT3A and sensitized these cells to growth inhibition mediated by the TkI. long-term exposure of CML k562 cells to ABL TkI such as dasatinib and nilotinib decreased the levels of miR-217 and increased the levels of DNMT1 and DNMT3A, as well as resulting in acquisition of TkI resistance. | |||
|
||||
Key Molecule: DNA (cytosine-5)-methyltransferase 3A (DNMT3A) | [17] | |||
Sensitive Disease | Chronic myelogenous Ph(+) leukemia [ICD-11: 2A20.1] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Nilotinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Bcr/Abl-expressing k562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
K562DR cells | Blood | Homo sapiens (Human) | CVCL_4V59 | |
K562NR cells | Blood | Homo sapiens (Human) | CVCL_4V63 | |
In Vivo Model | BALB/c nude mouse xenograft model | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | Forced expression of miR-217 inhibited expression of DNMT3A through a miR-217-binding site within the 3'-untranslated region of DNMT3A and sensitized these cells to growth inhibition mediated by the TkI. long-term exposure of CML k562 cells to ABL TkI such as dasatinib and nilotinib decreased the levels of miR-217 and increased the levels of DNMT1 and DNMT3A, as well as resulting in acquisition of TkI resistance. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: hsa-mir-370 | [79] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Sensitive Drug | Omacetaxine mepesuccinate | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | miR370/FoxM1 signaling pathway | Regulation | hsa05206 | |
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | miR-370 sensitized k562 cells to HHT by inducing apoptosis in part by downregulation of FoxM1 expression. | |||
|
||||
Key Molecule: Forkhead box protein M1 (FOXM1) | [79] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Omacetaxine mepesuccinate | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | miR370/FoxM1 signaling pathway | Regulation | hsa05206 | |
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
Flow cytometry assay | |||
Mechanism Description | miR-370 sensitized k562 cells to HHT by inducing apoptosis in part by downregulation of FoxM1 expression. |
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [80] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.T315I |
||
Resistant Drug | Ponatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Sanger sequencing assay | |||
Mechanism Description | Ponatinib was highly active in heavily pretreated patients with Ph-positive leukemias with resistance to tyrosine kinase inhibitors, including patients with the BCR-ABL T315I mutation, other mutations, or no mutations. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [80] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.T212R |
||
Resistant Drug | Ponatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Sanger sequencing assay | |||
Mechanism Description | Ponatinib was highly active in heavily pretreated patients with Ph-positive leukemias with resistance to tyrosine kinase inhibitors, including patients with the BCR-ABL T315I mutation, other mutations, or no mutations. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [80] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.Q252H |
||
Resistant Drug | Ponatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Sanger sequencing assay | |||
Mechanism Description | Ponatinib was highly active in heavily pretreated patients with Ph-positive leukemias with resistance to tyrosine kinase inhibitors, including patients with the BCR-ABL T315I mutation, other mutations, or no mutations. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [80] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.M244V |
||
Resistant Drug | Ponatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Sanger sequencing assay | |||
Mechanism Description | Ponatinib was highly active in heavily pretreated patients with Ph-positive leukemias with resistance to tyrosine kinase inhibitors, including patients with the BCR-ABL T315I mutation, other mutations, or no mutations. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [80] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.L387F |
||
Resistant Drug | Ponatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Sanger sequencing assay | |||
Mechanism Description | Ponatinib was highly active in heavily pretreated patients with Ph-positive leukemias with resistance to tyrosine kinase inhibitors, including patients with the BCR-ABL T315I mutation, other mutations, or no mutations. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [80] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.H396R |
||
Resistant Drug | Ponatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Sanger sequencing assay | |||
Mechanism Description | Ponatinib was highly active in heavily pretreated patients with Ph-positive leukemias with resistance to tyrosine kinase inhibitors, including patients with the BCR-ABL T315I mutation, other mutations, or no mutations. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [80] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.G250E |
||
Resistant Drug | Ponatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Sanger sequencing assay | |||
Mechanism Description | Ponatinib was highly active in heavily pretreated patients with Ph-positive leukemias with resistance to tyrosine kinase inhibitors, including patients with the BCR-ABL T315I mutation, other mutations, or no mutations. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [80] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F359V |
||
Resistant Drug | Ponatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Sanger sequencing assay | |||
Mechanism Description | Ponatinib was highly active in heavily pretreated patients with Ph-positive leukemias with resistance to tyrosine kinase inhibitors, including patients with the BCR-ABL T315I mutation, other mutations, or no mutations. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [80] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F359I |
||
Resistant Drug | Ponatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Sanger sequencing assay | |||
Mechanism Description | Ponatinib was highly active in heavily pretreated patients with Ph-positive leukemias with resistance to tyrosine kinase inhibitors, including patients with the BCR-ABL T315I mutation, other mutations, or no mutations. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [80] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F359C |
||
Resistant Drug | Ponatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Sanger sequencing assay | |||
Mechanism Description | Ponatinib was highly active in heavily pretreated patients with Ph-positive leukemias with resistance to tyrosine kinase inhibitors, including patients with the BCR-ABL T315I mutation, other mutations, or no mutations. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [80] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F317L |
||
Resistant Drug | Ponatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Sanger sequencing assay | |||
Mechanism Description | Ponatinib was highly active in heavily pretreated patients with Ph-positive leukemias with resistance to tyrosine kinase inhibitors, including patients with the BCR-ABL T315I mutation, other mutations, or no mutations. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [80] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E453K |
||
Resistant Drug | Ponatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Sanger sequencing assay | |||
Mechanism Description | Ponatinib was highly active in heavily pretreated patients with Ph-positive leukemias with resistance to tyrosine kinase inhibitors, including patients with the BCR-ABL T315I mutation, other mutations, or no mutations. | |||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [80] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E279K |
||
Resistant Drug | Ponatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Sanger sequencing assay | |||
Mechanism Description | Ponatinib was highly active in heavily pretreated patients with Ph-positive leukemias with resistance to tyrosine kinase inhibitors, including patients with the BCR-ABL T315I mutation, other mutations, or no mutations. | |||
|
||||
Key Molecule: hsa-miR-29a-3p | [15] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Ponatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Lin-CD34+CD38- cells | Bone | Homo sapiens (Human) | N.A. |
Lin-CD34-CD38- CML cells | Bone | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Annexin V assay | |||
Mechanism Description | The up-regulation of miR29a-3p observed in CML LSCs led to the down-regulation of its target TET2 and conferred TkI-resistance to CML LSCs in vitro. | |||
Key Molecule: hsa-miR-494-3p | [15] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Ponatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Lin-CD34+CD38- cells | Bone | Homo sapiens (Human) | N.A. |
Lin-CD34-CD38- CML cells | Bone | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Annexin V assay | |||
Mechanism Description | miR494-3p down-regulation in CML LSCs, leading to c-MYC up-regulation, was able to decrease TkI-induced apoptosis. | |||
Key Molecule: hsa-miR-660-5p | [15] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Ponatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Lin-CD34+CD38- cells | Bone | Homo sapiens (Human) | N.A. |
Lin-CD34-CD38- CML cells | Bone | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
qRT-PCR | |||
Experiment for Drug Resistance |
Annexin V assay | |||
Mechanism Description | The up-regulation of miR660-5p observed in CML LSCs led to the down-regulation of its target EPAS1 and conferred TkI-resistance to CML LSCs in vitro. | |||
|
||||
Key Molecule: Myc proto-oncogene protein (MYC) | [15] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Ponatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Lin-CD34+CD38- cells | Bone | Homo sapiens (Human) | N.A. |
Lin-CD34-CD38- CML cells | Bone | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Annexin V assay | |||
Mechanism Description | miR494-3p down-regulation in CML LSCs, leading to c-MYC up-regulation, was able to decrease TkI-induced apoptosis. | |||
Key Molecule: Hypoxia-inducible factor 2-alpha (EPAS1) | [15] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Ponatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Lin-CD34+CD38- cells | Bone | Homo sapiens (Human) | N.A. |
Lin-CD34-CD38- CML cells | Bone | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Annexin V assay | |||
Mechanism Description | The up-regulation of miR660-5p observed in CML LSCs led to the down-regulation of its target EPAS1 and conferred TkI-resistance to CML LSCs in vitro. | |||
Key Molecule: Methylcytosine dioxygenase TET2 (TET2) | [15] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Resistant Drug | Ponatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Lin-CD34+CD38- cells | Bone | Homo sapiens (Human) | N.A. |
Lin-CD34-CD38- CML cells | Bone | Homo sapiens (Human) | N.A. | |
Experiment for Molecule Alteration |
Western blot analysis | |||
Experiment for Drug Resistance |
Annexin V assay | |||
Mechanism Description | The up-regulation of miR29a-3p observed in CML LSCs led to the down-regulation of its target TET2 and conferred TkI-resistance to CML LSCs in vitro. | |||
Key Molecule: Cellular tumor antigen p53 (TP53) | [5] | |||
Resistant Disease | Chronic myelogenous leukemia [ICD-11: 2A20.3] | |||
Molecule Alteration | Missense mutation | p.V216M |
||
Resistant Drug | Ponatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | JAKT2/STAT signaling pathway | Activation | hsa04030 | |
RAF/KRAS/MEK signaling pathway | Activation | hsa04010 | ||
In Vitro Model | HL60 cells | Peripheral blood | Homo sapiens (Human) | CVCL_0002 |
U937 cells | Blood | Homo sapiens (Human) | CVCL_0007 | |
K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 | |
KCL-22 cells | Bone marrow | Homo sapiens (Human) | CVCL_2091 | |
Sup-B15 cells | Bone marrow | Homo sapiens (Human) | CVCL_0103 | |
HEL cells | Blood | Homo sapiens (Human) | CVCL_0001 | |
HMC-1.2 cells | Blood | Homo sapiens (Human) | CVCL_H205 | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Next-generation sequencing assay; Sanger Sequencing assay | |||
Mechanism Description | Po.tinib led to a decrease of ABL T315I positive transcripts from 47% before po.tinib treatment to 16% at the time of po.tinib resistance in this patient, suggesting that both TP53 and ABL mutations were present in the same clone and that the newly acquired TP53 mutation might have caused po.tinib resistance in this patient. | |||
Key Molecule: GTPase Nras (NRAS) | [4], [5] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.G12V |
||
Resistant Drug | Ponatinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | JAKT2/STAT signaling pathway | Activation | hsa04030 | |
RAF/KRAS/MEK signaling pathway | Activation | hsa04010 | ||
In Vitro Model | HL60 cells | Peripheral blood | Homo sapiens (Human) | CVCL_0002 |
U937 cells | Blood | Homo sapiens (Human) | CVCL_0007 | |
K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 | |
KCL-22 cells | Bone marrow | Homo sapiens (Human) | CVCL_2091 | |
Sup-B15 cells | Bone marrow | Homo sapiens (Human) | CVCL_0103 | |
HEL cells | Blood | Homo sapiens (Human) | CVCL_0001 | |
HMC-1.2 cells | Blood | Homo sapiens (Human) | CVCL_H205 | |
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Next-generation sequencing assay; Sanger Sequencing assay | |||
Mechanism Description | This mutation is well known for its effects on proliferation and its association with AML and MPN, suggesting that this variant might have been involved in the TkI resistance of this patient. |
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: Tyrosine-protein kinase JAK2 (JAK3) | [81] | |||
Resistant Disease | Polycythaemia vera [ICD-11: 2A20.4] | |||
Molecule Alteration | Missense mutation | p.V617F |
||
Resistant Drug | Ruxolitinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | JAK2 mutation confers resistance to Ruxolitinib. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: Tyrosine-protein kinase JAK2 (JAK3) | [6] | |||
Sensitive Disease | Primary myelofibrosis [ICD-11: 2A20.2] | |||
Molecule Alteration | Function | Inhibition |
||
Sensitive Drug | Ruxolitinib | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | In non-transplant candidates, conventional treatment for anemia includes androgens, prednisone, thalidomide, and danazol; for symptomatic splenomegaly, hydroxyurea and ruxolitinib; and for constitutional symptoms, ruxolitinib. Fedratinib, another JAK2 inhibitor, has now been FDA-approved for use in ruxolitinib failures. | |||
|
||||
Key Molecule: Granulocyte colony-stimulating factor receptor (CSF3R) | [82] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.T618I (c.1853C>T) |
||
Sensitive Drug | Ruxolitinib | |||
Experimental Note | Discovered Using In-vivo Testing Model | |||
In Vivo Model | BALB/C nude mouse xenograft model | Mus musculus |
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: Multidrug resistance protein 1 (ABCB1) | [83] | |||
Resistant Disease | Chronic myelogenous leukemia [ICD-11: 2A20.3] | |||
Molecule Alteration | Expression | Up-regulation |
||
Resistant Drug | Temozolomide | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Inhibition | hsa04210 | |
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
K562-VP16 cells | Blood | Homo sapiens (Human) | CVCL_0004 | |
Experiment for Molecule Alteration |
Real-time PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | In the chemosensitive MDR1-negative parental cell line k562 10 ug/ml temozolomide resulted in pronounced cell death with only 47.1% surviving 48 h compared with the control. In contrast, in the highly MDR1-expressing resistant subline k562-VP16, cell death was significantly lower after exposure to temozolomide with 73.4% surviving 48 h (P = 0.002). Addition of a nontoxic dose of the MDR1-modulator cyclosporine A (1 uM) to temozolomide resulted in a trend towards restoration of chemosensitivity in the resistant MDR1-expressing cell line. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: Multidrug resistance protein 1 (ABCB1) | [83] | |||
Sensitive Disease | Chronic myelogenous leukemia [ICD-11: 2A20.3] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Temozolomide | |||
Experimental Note | Identified from the Human Clinical Data | |||
Cell Pathway Regulation | Cell apoptosis | Activation | hsa04210 | |
In Vitro Model | K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 |
K562-VP16 cells | Blood | Homo sapiens (Human) | CVCL_0004 | |
Experiment for Molecule Alteration |
Real-time PCR | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | In the chemosensitive MDR1-negative parental cell line k562 10 ug/ml temozolomide resulted in pronounced cell death with only 47.1% surviving 48 h compared with the control. In contrast, in the highly MDR1-expressing resistant subline k562-VP16, cell death was significantly lower after exposure to temozolomide with 73.4% surviving 48 h (P = 0.002). Addition of a nontoxic dose of the MDR1-modulator cyclosporine A (1 uM) to temozolomide resulted in a trend towards restoration of chemosensitivity in the resistant MDR1-expressing cell line. |
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: Multidrug resistance protein 1 (ABCB1) | [18] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Expression | Down-regulation |
||
Sensitive Drug | Vinblastine | |||
Experimental Note | Revealed Based on the Cell Line Data | |||
In Vitro Model | NCI-H460 cells | Lung | Homo sapiens (Human) | CVCL_0459 |
K562 cells | Blood | Homo sapiens (Human) | CVCL_0004 | |
HEK293 cells | Kidney | Homo sapiens (Human) | CVCL_0045 | |
K562-R cells | Pleural effusion | Homo sapiens (Human) | CVCL_5950 | |
NCI-H460/VBL cells | Bone marrow | Homo sapiens (Human) | CVCL_0459 | |
In Vivo Model | SCID beige mice | Mus musculus | ||
Experiment for Molecule Alteration |
Western blotting analysis | |||
Experiment for Drug Resistance |
MTT assay | |||
Mechanism Description | In ABCB1-overexpressing cell lines, HG-829 significantly enhanced cytotoxicity to daunorubicin, paclitaxel, vinblastine, vincristine, and etoposide. Coadministration of HG-829 fully restored in vivo antitumor activity of daunorubicin in mice without added toxicity. Functional assays showed that HG-829 is not a Pgp substrate or competitive inhibitor of Pgp-mediated drug efflux but rather acts as a noncompetitive modulator of P-glycoprotein transport function. |
Clinical Trial Drug(s)
1 drug(s) in total
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: Tyrosine-protein kinase JAK2 (JAK3) | [84] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.V617F (c.1849G>T) |
||
Sensitive Drug | Ropeginterferon alfa-2b | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | HEL cells | Blood | Homo sapiens (Human) | CVCL_0001 |
UKE-1 cells | Peripheral blood | Homo sapiens (Human) | CVCL_0104 | |
Experiment for Drug Resistance |
Trypan blue staining assay |
Investigative Drug(s)
2 drug(s) in total
Drug Sensitivity Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: Tyrosine-protein kinase JAK2 (JAK3) | [85] | |||
Sensitive Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.V617F (c.1849G>T) |
||
Sensitive Drug | Peginterferon alfa-2a | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vitro Model | Bone marrow | . | ||
Mechanism Description | The missense mutation p.V617F (c.1849G>T) in gene JAK2 cause the sensitivity of Peginterferon alfa-2a by unusual activation of pro-survival pathway |
Drug Resistance Data Categorized by Their Corresponding Mechanisms | ||||
|
||||
Key Molecule: BCR-ABL1 e8a2 variant (BCR-ABL1) | [42] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.L324Q |
||
Resistant Drug | Tyrosine kinase inhibitor | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Pyrosequencing analysis | |||
Experiment for Drug Resistance |
Progression-free survival assay; Overall survival assay | |||
Mechanism Description | Imatinib resistance in chronic myeloid leukemia (CML) is commonly due to BCR-ABL kinase domain mutations (kDMs). | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [80] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.M351T |
||
Resistant Drug | Tyrosine kinase inhibitor | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Sanger sequencing assay | |||
Mechanism Description | Ponatinib was highly active in heavily pretreated patients with Ph-positive leukemias with resistance to tyrosine kinase inhibitors, including patients with the BCR-ABL T315I mutation, other mutations, or no mutations. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [9], [86] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F317L |
||
Resistant Drug | Tyrosine kinase inhibitor | |||
Experimental Note | Identified from the Human Clinical Data | |||
In Vivo Model | A retrospective survey in conducting clinical studies | Homo sapiens | ||
Experiment for Molecule Alteration |
Direct sequencing assay | |||
Mechanism Description | We confirmed the high frequency of SFks involvement in Tyrosine kinase inhibitor-resistant CML (52% of the cases) and even further in progressive disease and blast crises (60% of the cases). The SFks deregulation is also observed in patients harboring BCR-ABL mutations. In T315I and F317L mutated patients, CML-resistance appears to be promoted by SFks kinase protein reactivation once the BCR-ABL mutated clone has decreased on Omacetaxine. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [86] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.M244V |
||
Resistant Drug | Tyrosine kinase inhibitor | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | There are several identified mechanisms of resistance to TkIs. The presence of ABL kinase domain point mutation, which could be detected by molecular methods is one of them. ABL mutation was detected in 19 (31,6%) patients. In four cases with detected mutation the disease has progressed to blast crisis. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [86] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.V299L |
||
Resistant Drug | Tyrosine kinase inhibitor | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | There are several identified mechanisms of resistance to TkIs. The presence of ABL kinase domain point mutation, which could be detected by molecular methods is one of them. ABL mutation was detected in 19 (31,6%) patients. In four cases with detected mutation the disease has progressed to blast crisis. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [86] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.V289A |
||
Resistant Drug | Tyrosine kinase inhibitor | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | There are several identified mechanisms of resistance to TkIs. The presence of ABL kinase domain point mutation, which could be detected by molecular methods is one of them. ABL mutation was detected in 19 (31,6%) patients. In four cases with detected mutation the disease has progressed to blast crisis. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [86] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.T315I |
||
Resistant Drug | Tyrosine kinase inhibitor | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | There are several identified mechanisms of resistance to TkIs. The presence of ABL kinase domain point mutation, which could be detected by molecular methods is one of them. ABL mutation was detected in 19 (31,6%) patients. In four cases with detected mutation the disease has progressed to blast crisis. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [86] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.R220H |
||
Resistant Drug | Tyrosine kinase inhibitor | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | There are several identified mechanisms of resistance to TkIs. The presence of ABL kinase domain point mutation, which could be detected by molecular methods is one of them. ABL mutation was detected in 19 (31,6%) patients. In four cases with detected mutation the disease has progressed to blast crisis. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [86] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.Q252H |
||
Resistant Drug | Tyrosine kinase inhibitor | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | There are several identified mechanisms of resistance to TkIs. The presence of ABL kinase domain point mutation, which could be detected by molecular methods is one of them. ABL mutation was detected in 19 (31,6%) patients. In four cases with detected mutation the disease has progressed to blast crisis. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [86] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.L384M |
||
Resistant Drug | Tyrosine kinase inhibitor | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | There are several identified mechanisms of resistance to TkIs. The presence of ABL kinase domain point mutation, which could be detected by molecular methods is one of them. ABL mutation was detected in 19 (31,6%) patients. In four cases with detected mutation the disease has progressed to blast crisis. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [86] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.H396R |
||
Resistant Drug | Tyrosine kinase inhibitor | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | There are several identified mechanisms of resistance to TkIs. The presence of ABL kinase domain point mutation, which could be detected by molecular methods is one of them. ABL mutation was detected in 19 (31,6%) patients. In four cases with detected mutation the disease has progressed to blast crisis. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [86] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.G250E |
||
Resistant Drug | Tyrosine kinase inhibitor | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | There are several identified mechanisms of resistance to TkIs. The presence of ABL kinase domain point mutation, which could be detected by molecular methods is one of them. ABL mutation was detected in 19 (31,6%) patients. In four cases with detected mutation the disease has progressed to blast crisis. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [86] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F359I |
||
Resistant Drug | Tyrosine kinase inhibitor | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | There are several identified mechanisms of resistance to TkIs. The presence of ABL kinase domain point mutation, which could be detected by molecular methods is one of them. ABL mutation was detected in 19 (31,6%) patients. In four cases with detected mutation the disease has progressed to blast crisis. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [86] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.F311L |
||
Resistant Drug | Tyrosine kinase inhibitor | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | There are several identified mechanisms of resistance to TkIs. The presence of ABL kinase domain point mutation, which could be detected by molecular methods is one of them. ABL mutation was detected in 19 (31,6%) patients. In four cases with detected mutation the disease has progressed to blast crisis. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [86] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.E282K |
||
Resistant Drug | Tyrosine kinase inhibitor | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | There are several identified mechanisms of resistance to TkIs. The presence of ABL kinase domain point mutation, which could be detected by molecular methods is one of them. ABL mutation was detected in 19 (31,6%) patients. In four cases with detected mutation the disease has progressed to blast crisis. | |||
Key Molecule: Tyrosine-protein kinase ABL1 (ABL1) | [86] | |||
Resistant Disease | Chronic myeloid leukemia [ICD-11: 2A20.0] | |||
Molecule Alteration | Missense mutation | p.A399T |
||
Resistant Drug | Tyrosine kinase inhibitor | |||
Experimental Note | Identified from the Human Clinical Data | |||
Mechanism Description | There are several identified mechanisms of resistance to TkIs. The presence of ABL kinase domain point mutation, which could be detected by molecular methods is one of them. ABL mutation was detected in 19 (31,6%) patients. In four cases with detected mutation the disease has progressed to blast crisis. |
References
If you find any error in data or bug in web service, please kindly report it to Dr. Sun and Dr. Zhang.