1
|
Nguyen T, Harama D, Tamai M, Kagami K, Komatsu C, Kasai S, Akahane K, Goi K, Inukai T. Synergistic effect of asciminib with reduced doses of ponatinib in human Ph + myeloid leukemia with the T315M mutation. Int J Hematol 2025:10.1007/s12185-025-03981-7. [PMID: 40208408 DOI: 10.1007/s12185-025-03981-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/27/2025] [Accepted: 03/27/2025] [Indexed: 04/11/2025]
Abstract
In Philadelphia chromosome-positive (Ph +) leukemia, substitution of threonine at the 315 position of BCR::ABL1 with isoleucine (T315I) induces severe resistance to tyrosine kinase inhibitors (TKIs). Of clinical importance, the substitution of the baseline T315I mutation by methionine (I315M) was reported in a Ph + leukemia patient treated with ponatinib. The resultant T315M mutation induces severe TKI-resistance in a murine Ba/F3 model. Asciminib, an allosteric inhibitor of BCR::ABL1, is reportedly active in ponatinib-resistant patients with the T315I mutation. Although asciminib alone is not active in a murine Ba/F3 model with the T315M mutation, asciminib and ponatinib show synergistic activities. In the present study, we introduced the T315M mutation into the intrinsic BCR::ABL1 gene of two Ph + myeloid and one Ph + lymphoid leukemia cell lines using the CRISPR/Cas9 system to directly verify the utility of the combined asciminib and ponatinib in human models. All three T315M-acquired sublines were more resistant to TKIs including ponatinib than T315I-acquired sublines. Notably, asciminib exhibited a stronger synergistic effect with reduced doses of ponatinib in the T315M-acquired sublines of two myeloid cell lines, but not in the lymphoid cell line. This indicates that the combination of ponatinib and asciminib may have a clinical utility in human Ph + myeloid leukemia.
Collapse
Affiliation(s)
- Thao Nguyen
- Global Leukemia Cell-Line Assembly Network and Department of Pediatrics, School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409 - 3898, Japan
| | - Daisuke Harama
- Global Leukemia Cell-Line Assembly Network and Department of Pediatrics, School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409 - 3898, Japan
| | - Minori Tamai
- Global Leukemia Cell-Line Assembly Network and Department of Pediatrics, School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409 - 3898, Japan
| | - Keiko Kagami
- Global Leukemia Cell-Line Assembly Network and Department of Pediatrics, School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409 - 3898, Japan
| | - Chiaki Komatsu
- Global Leukemia Cell-Line Assembly Network and Department of Pediatrics, School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409 - 3898, Japan
| | - Shin Kasai
- Global Leukemia Cell-Line Assembly Network and Department of Pediatrics, School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409 - 3898, Japan
| | - Koshi Akahane
- Global Leukemia Cell-Line Assembly Network and Department of Pediatrics, School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409 - 3898, Japan
| | - Kumiko Goi
- Global Leukemia Cell-Line Assembly Network and Department of Pediatrics, School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409 - 3898, Japan
| | - Takeshi Inukai
- Global Leukemia Cell-Line Assembly Network and Department of Pediatrics, School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409 - 3898, Japan.
| |
Collapse
|
2
|
Nguyen T, Tamai M, Fujisawa S, Nagamachi A, Kagami K, Komatsu C, Iwamoto S, Inaba T, Teshima T, Inukai T. Utility of the Base Editing System for Introducing Drug-Resistant Gene Mutations Into Human Leukemia Cellular Models. Cureus 2025; 17:e81889. [PMID: 40342439 PMCID: PMC12060998 DOI: 10.7759/cureus.81889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2025] [Indexed: 05/11/2025] Open
Abstract
Background Recent genomic analyses of poor prognostic and relapsed leukemia have revealed the involvement of diverse gene mutations in treatment resistance. These gene mutations are classified into two groups: mutations involving resistance to specific agents such as the BCR::ABL1 fusion gene mutations (typically T315I mutation) in tyrosine kinase inhibitor (TKI) resistance and those involving the resistance to diverse therapeutic modalities such as the TP53 gene mutations. In the latter type, although their associations with drug resistance have been clinically demonstrated, the direct association with resistance to each therapeutic modality remains to be fully elucidated. To overcome treatment resistance induced by these gene mutations, appropriate leukemic cellular models are urgently required. Using the cytidine base editing (CBE) system, we introduced two types of mutations through C-to-T transition into human leukemia cell lines and evaluated their significance in the drug sensitivities. Methods We applied the CBE system to introduce the T315I (ACT to ATT) mutation of the BCR::ABL1 fusion gene in a human Philadelphia chromosome-positive leukemia cell line and to introduce the T125M (ACG to ATG) mutation of the TP53 gene in a human B-cell precursor acute lymphoblastic leukemia (BCP-ALL) cell line. Results We first confirmed an introduction of the T315I mutation in one of four BCR::ABL1 alleles as a result of base editing in the obtained TKI-resistant subline. We also identified the additional C-to-T transition at adjacent codon 314 (ATC), which resulted in a silent mutation, in the same allele. We next confirmed that the obtained subline acquired the T125M mutation of the TP53 gene without additional C-to-T transition. In the T125M subline, transcriptional activities of the p53 protein were disrupted and the sensitivities to diverse chemotherapeutic drugs and irradiation were reduced. Conclusion Our observations demonstrated the utility of the CBE system for introducing specific nucleotide transitions into human leukemia cell lines.
Collapse
Affiliation(s)
- Thao Nguyen
- Pediatrics, University of Yamanashi, Chuo, JPN
| | - Minori Tamai
- Pediatrics, Global Leukemia Cell-Line Assembly Network, University of Yamanashi, Chuo, JPN
| | - Shinichi Fujisawa
- Laboratory and Transfusion Medicine, Hokkaido University Hospital, Sapporo, JPN
| | - Akiko Nagamachi
- Molecular Oncology and Leukemia Program Project, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, JPN
| | - Keiko Kagami
- Global Leukemia Cell-Line Assembly Network, University of Yamanashi, Chuo, JPN
| | - Chiaki Komatsu
- Global Leukemia Cell-Line Assembly Network, University of Yamanashi, Chuo, JPN
| | - Shotaro Iwamoto
- Pediatrics, Mie University Graduate School of Medicine, Tsu, JPN
| | - Toshiya Inaba
- Molecular Oncology and Leukemia Program Project, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, JPN
| | - Takanori Teshima
- Laboratory and Transfusion Medicine, Hokkaido University Hospital, Sapporo, JPN
| | - Takeshi Inukai
- Pediatrics, Global Leukemia Cell-Line Assembly Network, University of Yamanashi, Chuo, JPN
| |
Collapse
|
3
|
Nguyen TT, Tamai M, Harama D, Kagami K, Komatsu C, Kasai S, Akahane K, Goi K, Inukai T. Sensitivity to Tyrosine Kinase Inhibitors in a Human Philadelphia Chromosome-Positive (Ph+) Leukemia Model With the T315I-Inclusive Compound Mutation. Cureus 2024; 16:e76538. [PMID: 39872583 PMCID: PMC11772067 DOI: 10.7759/cureus.76538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2024] [Indexed: 01/30/2025] Open
Abstract
The T315I-inclusive compound mutation, the multiple mutations including the T315I mutation on the same BCR::ABL1 gene, confers resistance to diverse tyrosine kinase inhibitors (TKIs). Development of the F311I/T315I compound mutation has been reported in chronic myeloid leukemia patients who sequentially showed clinical resistance to imatinib and dasatinib. The establishment of a human leukemia model with the T315I-inclusive compound mutation remains an experimental challenge. Here, we introduced the F311I/T315I compound mutation into the intrinsic BCR::ABL1 gene of a human TKI-sensitive Philadelphia chromosome-positive leukemia cell line via homologous recombination using the CRISPR/Cas9 system and obtained three types of sublines: the F311I mutation alone, the T315I mutation alone, and the F311I/T315I compound mutation. The F311I subline was sensitive to dasatinib but moderately resistant to imatinib and nilotinib, while the T315I subline and the F311I/T315I subline were highly resistant to these TKIs. Notably, the T315I subline and the F311I/T315I subline were sensitive to therapeutic concentrations of ponatinib, although more resistant than the F311I subline. Moreover, the T315 subline and the F311I/T315 subline were sensitive to asciminib at therapeutic concentration, as was the F311I subline. This is the first human leukemia model in which the impact of the T315I-inclusive compound mutation on TKI sensitivity was directly confirmed.
Collapse
Affiliation(s)
- Thao T Nguyen
- Department of Pediatrics, University of Yamanashi, Chuo, JPN
| | - Minori Tamai
- Department of Pediatrics, University of Yamanashi, Chuo, JPN
| | - Daisuke Harama
- Department of Pediatrics, University of Yamanashi, Chuo, JPN
| | - Keiko Kagami
- Department of Pediatrics, University of Yamanashi, Chuo, JPN
| | - Chiaki Komatsu
- Department of Pediatrics, University of Yamanashi, Chuo, JPN
| | - Shin Kasai
- Department of Pediatrics, University of Yamanashi, Chuo, JPN
| | - Koshi Akahane
- Department of Pediatrics, University of Yamanashi, Chuo, JPN
| | - Kumiko Goi
- Department of Pediatrics, University of Yamanashi, Chuo, JPN
| | - Takeshi Inukai
- Department of Pediatrics, University of Yamanashi, Chuo, JPN
| |
Collapse
|
4
|
Okamoto N, Yagi K, Imawaka S, Takaoka M, Aizawa F, Niimura T, Goda M, Miyata K, Kawada K, Izawa‐Ishizawa Y, Sakaguchi S, Ishizawa K. Asciminib, a novel allosteric inhibitor of BCR-ABL1, shows synergistic effects when used in combination with imatinib with or without drug resistance. Pharmacol Res Perspect 2024; 12:e1214. [PMID: 39031848 PMCID: PMC11191601 DOI: 10.1002/prp2.1214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/25/2024] [Accepted: 04/15/2024] [Indexed: 07/22/2024] Open
Abstract
In the treatment of chronic myeloid leukemia (CML), resistance to BCR-ABL inhibitors makes it difficult to continue treatment and is directly related to life expectancy. Therefore, asciminib was introduced to the market as a useful drug for overcoming drug resistance. While combining molecular targeted drugs is useful to avoid drug resistance, the new BCR-ABL inhibitor asciminib and conventional BCR-ABL inhibitors should be used as monotherapy in principle. Therefore, we investigated the synergistic effect and mechanism of the combination of asciminib and imatinib. We generated imatinib-resistant cells using the human CML cell line K562, examined the effects of imatinib and asciminib exposure on cell survival using the WST-8 assay, and comprehensively analyzed genetic variation related to drug resistance using RNA-seq and real-time PCR. A synergistic effect was observed when imatinib and asciminib were combined with or without imatinib resistance. Three genes, GRRP1, ESPN, and NOXA1, were extracted as the sites of action of asciminib. Asciminib in combination with BCR-ABL inhibitors may improve the therapeutic efficacy of conventional BCR-ABL inhibitors and prevent the development of resistance. Its dosage may be effective even at minimal doses that do not cause side effects. Further verification of this mechanism of action is needed. Additionally, cross-resistance between BCR-ABL inhibitors and asciminib may occur, which needs to be clarified through further validation as soon as possible.
Collapse
MESH Headings
- Imatinib Mesylate/pharmacology
- Humans
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/genetics
- Drug Synergism
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Protein Kinase Inhibitors/pharmacology
- Cell Survival/drug effects
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Agents/pharmacology
- Niacinamide/analogs & derivatives
- Pyrazoles
Collapse
Affiliation(s)
- Naoki Okamoto
- Department of Clinical Pharmacology and TherapeuticsTokushima University Graduate School of Biomedical SciencesTokushimaJapan
- Department of PharmacyTokushima University HospitalTokushimaJapan
| | - Kenta Yagi
- Department of Clinical Pharmacology and TherapeuticsTokushima University Graduate School of Biomedical SciencesTokushimaJapan
- Clinical Research Center for Developmental TherapeuticsTokushima University HospitalTokushimaJapan
| | - Sayaka Imawaka
- Department of Clinical Pharmacology and TherapeuticsTokushima University Graduate School of Biomedical SciencesTokushimaJapan
| | - Mayu Takaoka
- Department of Clinical Pharmacology and TherapeuticsTokushima University Graduate School of Biomedical SciencesTokushimaJapan
| | - Fuka Aizawa
- Department of Clinical Pharmacology and TherapeuticsTokushima University Graduate School of Biomedical SciencesTokushimaJapan
- Department of PharmacyTokushima University HospitalTokushimaJapan
| | - Takahiro Niimura
- Department of Clinical Pharmacology and TherapeuticsTokushima University Graduate School of Biomedical SciencesTokushimaJapan
- Clinical Research Center for Developmental TherapeuticsTokushima University HospitalTokushimaJapan
| | - Mitsuhiro Goda
- Department of Clinical Pharmacology and TherapeuticsTokushima University Graduate School of Biomedical SciencesTokushimaJapan
- Department of PharmacyTokushima University HospitalTokushimaJapan
| | - Koji Miyata
- Department of Clinical Pharmacology and TherapeuticsTokushima University Graduate School of Biomedical SciencesTokushimaJapan
| | - Kei Kawada
- Department of Clinical Pharmacology and TherapeuticsTokushima University Graduate School of Biomedical SciencesTokushimaJapan
- Department of Clinical Pharmacy Practice PedagogyTokushima University Graduate School of Biomedical SciencesTokushimaJapan
| | - Yuki Izawa‐Ishizawa
- Department of Clinical Pharmacology and TherapeuticsTokushima University Graduate School of Biomedical SciencesTokushimaJapan
- Department of General MedicineTaoka HospitalTokushimaJapan
| | - Satoshi Sakaguchi
- Clinical Research Center for Developmental TherapeuticsTokushima University HospitalTokushimaJapan
- Department of Respiratory Medicine and RheumatologyTokushima University Graduate School of Biomedical SciencesTokushimaJapan
| | - Keisuke Ishizawa
- Department of Clinical Pharmacology and TherapeuticsTokushima University Graduate School of Biomedical SciencesTokushimaJapan
- Department of PharmacyTokushima University HospitalTokushimaJapan
- Clinical Research Center for Developmental TherapeuticsTokushima University HospitalTokushimaJapan
| |
Collapse
|
5
|
Nguyen T, Aida T, Iijima‐Yamashita Y, Tamai M, Nagamachi A, Kagami K, Komatsu C, Kasai S, Akahane K, Goi K, Inaba T, Sanada M, Inukai T. Application of prime editing system to introduce TP53 R248Q hotspot mutation in acute lymphoblastic leukemia cell line. Cancer Sci 2024; 115:1924-1935. [PMID: 38549229 PMCID: PMC11145152 DOI: 10.1111/cas.16162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 06/04/2024] Open
Abstract
In childhood acute lymphoblastic leukemia (ALL), TP53 gene mutation is associated with chemoresistance in a certain population of relapsed cases. To directly verify the association of TP53 gene mutation with chemoresistance of relapsed childhood ALL cases and improve their prognosis, the development of appropriate human leukemia models having TP53 mutation in the intrinsic gene is required. Here, we sought to introduce R248Q hotspot mutation into the intrinsic TP53 gene in an ALL cell line, 697, by applying a prime editing (PE) system, which is a versatile genome editing technology. The PE2 system uses an artificial fusion of nickase Cas9 and reverse-transcriptase to directly place new genetic information into a target site through a reverse transcriptase template in the prime editing guide RNA (pegRNA). Moreover, in the advanced PE3b system, single guide RNA (sgRNA) matching the edited sequence is also introduced to improve editing efficiency. The initially obtained MDM2 inhibitor-resistant PE3b-transfected subline revealed disrupted p53 transactivation activity, reduced p53 target gene expression, and acquired resistance to chemotherapeutic agents and irradiation. Although the majority of the subline acquired the designed R248Q and adjacent silent mutations, the insertion of the palindromic sequence in the scaffold hairpin structure of pegRNA and the overlap of the original genomic DNA sequence were frequently observed. Targeted next-generation sequencing reconfirmed frequent edit errors in both PE2 and PE3b-transfected 697 cells, and it revealed frequent successful edits in HEK293T cells. These observations suggest a requirement for further modification of the PE2 and PE3b systems for accurate editing in leukemic cells.
Collapse
Affiliation(s)
- Thao Nguyen
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| | - Tomomi Aida
- McGovern Institute for Brain ResearchMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Yuka Iijima‐Yamashita
- Department of Advanced DiagnosisClinical Research Center, NHO Nagoya Medical CenterNagoyaJapan
| | - Minori Tamai
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| | - Akiko Nagamachi
- Department of Molecular Oncology and Leukemia Program ProjectResearch Institute for Radiation Biology and Medicine, Hiroshima UniversityHigashihiroshimaJapan
| | - Keiko Kagami
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| | - Chiaki Komatsu
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| | - Shin Kasai
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| | - Koshi Akahane
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| | - Kumiko Goi
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| | - Toshiya Inaba
- Department of Molecular Oncology and Leukemia Program ProjectResearch Institute for Radiation Biology and Medicine, Hiroshima UniversityHigashihiroshimaJapan
| | - Masashi Sanada
- Department of Advanced DiagnosisClinical Research Center, NHO Nagoya Medical CenterNagoyaJapan
| | - Takeshi Inukai
- Department of Pediatrics, School of MedicineUniversity of YamanashiChuoJapan
| |
Collapse
|
6
|
Nussinov R, Jang H. Direct K-Ras Inhibitors to Treat Cancers: Progress, New Insights, and Approaches to Treat Resistance. Annu Rev Pharmacol Toxicol 2024; 64:231-253. [PMID: 37524384 DOI: 10.1146/annurev-pharmtox-022823-113946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Here we discuss approaches to K-Ras inhibition and drug resistance scenarios. A breakthrough offered a covalent drug against K-RasG12C. Subsequent innovations harnessed same-allele drug combinations, as well as cotargeting K-RasG12C with a companion drug to upstream regulators or downstream kinases. However, primary, adaptive, and acquired resistance inevitably emerge. The preexisting mutation load can explain how even exceedingly rare mutations with unobservable effects can promote drug resistance, seeding growth of insensitive cell clones, and proliferation. Statistics confirm the expectation that most resistance-related mutations are in cis, pointing to the high probability of cooperative, same-allele effects. In addition to targeted Ras inhibitors and drug combinations, bifunctional molecules and innovative tri-complex inhibitors to target Ras mutants are also under development. Since the identities and potential contributions of preexisting and evolving mutations are unknown, selecting a pharmacologic combination is taxing. Collectively, our broad review outlines considerations and provides new insights into pharmacology and resistance.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, Maryland, USA;
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, Maryland, USA;
| |
Collapse
|
7
|
Andreescu M. Recent Advances in Serum Biomarkers for Cardiological Risk Stratification and Insight into the Cardiac Management of the Patients With Hematological Malignancies Treated With Targeted Therapy. Cureus 2023; 15:e49696. [PMID: 38033434 PMCID: PMC10688222 DOI: 10.7759/cureus.49696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2023] [Indexed: 12/02/2023] Open
Abstract
Cardiovascular diseases (CVD) have emerged as a common and serious complication of cancer treatment, particularly in patients undergoing cardiotoxic therapies. Over the last few years, the medical community has become increasingly aware of the potential for cardiotoxicity resulting from cancer treatments involving chemotherapy, targeted therapies, and radiation therapy. This recognition is due to the significant risk of morbidity and mortality in cancer patients and survivors resulting from such treatment-induced cardiovascular damage. While the cardiotoxic effects of chemotherapy and targeted therapy have been discussed in medical literature, only a limited number of studies have explored the role of serum biomarkers in cardiological risk stratification. In recent years, serum biomarkers have emerged as a valuable tool for assessing and managing cardiotoxicity in patients with hematological malignancies. This review article provides a summary of the current state of knowledge on the usefulness of biomarkers in managing cardiotoxicity resulting from different targeted therapies throughout the cancer care continuum. Although cardiac biomarkers have demonstrated potential in identifying subclinical cardiotoxicity and tracking the response to cardioprotective treatments, further research is necessary to determine optimal biomarkers and surveillance strategies. The incorporation of cardiac biomarkers into clinical practice in patients undergoing targeted therapies could potentially lead to improved long-term cardiovascular outcomes in cancer patients and survivors.
Collapse
Affiliation(s)
- Mihaela Andreescu
- Department of Hematology, Colentina Clinical Hospital, Bucharest, ROU
- Department of Clinical Sciences, Hematology, Faculty of Medicine, Titu Maiorescu University of Bucharest, Bucharest, ROU
| |
Collapse
|
8
|
Hiroki H, Ishii Y, Piao J, Namikawa Y, Masutani M, Honda H, Akahane K, Inukai T, Morio T, Takagi M. Targeting Poly(ADP)ribose polymerase in BCR/ABL1-positive cells. Sci Rep 2023; 13:7588. [PMID: 37165001 PMCID: PMC10172294 DOI: 10.1038/s41598-023-33852-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 04/20/2023] [Indexed: 05/12/2023] Open
Abstract
BCR/ABL1 causes dysregulated cell proliferation and is responsible for chronic myelogenous leukemia (CML) and Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph1-ALL). In addition to the deregulatory effects of its kinase activity on cell proliferation, BCR/ABL1 induces genomic instability by downregulating BRCA1. PARP inhibitors (PARPi) effectively induce cell death in BRCA-defective cells. Therefore, PARPi are expected to inhibit growth of CML and Ph1-ALL cells showing downregulated expression of BRCA1. Here, we show that PARPi effectively induced cell death in BCR/ABL1 positive cells and suppressed colony forming activity. Prevention of BCR/ABL1-mediated leukemogenesis by PARP inhibition was tested in two in vivo models: wild-type mice that had undergone hematopoietic cell transplantation with BCR/ABL1-transduced cells, and a genetic model constructed by crossing Parp1 knockout mice with BCR/ABL1 transgenic mice. The results showed that a PARPi, olaparib, attenuates BCR/ABL1-mediated leukemogenesis. One possible mechanism underlying PARPi-dependent inhibition of leukemogenesis is increased interferon signaling via activation of the cGAS/STING pathway. This is compatible with the use of interferon as a first-line therapy for CML. Because tyrosine kinase inhibitor (TKI) monotherapy does not completely eradicate leukemic cells in all patients, combined use of PARPi and a TKI is an attractive option that may eradicate CML stem cells.
Collapse
Affiliation(s)
- Haruka Hiroki
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Yuko Ishii
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Jinhua Piao
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Yui Namikawa
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Mitsuko Masutani
- Department of Molecular and Genomic Biomedicine, Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, 852-8523, Nagasaki, Japan
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Hiroaki Honda
- Field of Human Disease Models, Major in Advanced Life Sciences and Medicine, Institute of Laboratory Animals, Tokyo Women's Medical University, Tokyo, Japan
| | - Koshi Akahane
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Takeshi Inukai
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Tomohiro Morio
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Masatoshi Takagi
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8519, Japan.
| |
Collapse
|
9
|
Nguyen TTT, Tanaka Y, Sanada M, Hosaka M, Tamai M, Kagami K, Komatsu C, Somazu S, Harama D, Kasai S, Watanabe A, Akahane K, Goi K, Inukai T. CRISPR/Cas9-Mediated Induction of Relapse-Specific NT5C2 and PRPS1 Mutations Confers Thiopurine Resistance as a Relapsed Lymphoid Leukemia Model. Mol Pharmacol 2023; 103:199-210. [PMID: 36669880 DOI: 10.1124/molpharm.122.000546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 12/13/2022] [Accepted: 01/04/2023] [Indexed: 01/21/2023] Open
Abstract
6-Mercaptopurine (6-MP) is a key component in maintenance therapy for childhood acute lymphoblastic leukemia (ALL). Recent next-generation sequencing analysis of childhood ALL clarified the emergence of the relapse-specific mutations of the NT5C2 and PRPS1 genes, which are involved in thiopurine metabolism. In this scenario, minor clones of leukemia cells could acquire the 6-MP-resistant phenotype as a result of the NT5C2 or PRPS1 mutation during chemotherapy (including 6-MP treatment) and confer disease relapse after selective expansion. Thus, to establish new therapeutic modalities overcoming 6-MP resistance in relapsed ALL, human leukemia models with NT5C2 and PRPS1 mutations in the intrinsic genes are urgently required. Here, mimicking the initiation process of the above clinical course, we sought to induce two relapse-specific hotspot mutations (R39Q mutation of the NT5C2 gene and S103N mutation of the PRPS1 gene) into a human lymphoid leukemia cell line by homologous recombination (HR) using the CRISPR/Cas9 system. After 6-MP selection of the cells transfected with Cas9 combined with single-guide RNA and donor DNA templates specific for either of those two mutations, we obtained the sublines with the intended NT5C2-R39Q and PRPS1-S103N mutation as a result of HR. Moreover, diverse in-frame small insertion/deletions were also confirmed in the 6-MP-resistant sublines at the target sites of the NT5C2 and PRPS1 genes as a result of nonhomologous end joining. These sublines are useful for molecular pharmacological evaluation of the NT5C2 and PRPS1 gene mutations in the 6-MP sensitivity and development of therapy overcoming the thiopurine resistance of leukemia cells. SIGNIFICANCE STATEMENT: Mimicking the initiation process of relapse-specific mutations of the NT5C2 and PRPS1 genes in childhood acute lymphoblastic leukemia treated with 6-mercaptopurine (6-MP), this study sought to introduce NT5C2-R39Q and PRPS1-S103N mutations into a human lymphoid leukemia cell line by homologous recombination using the CRISPR/Cas9 system. In the resultant 6-MP-resistant sublines, the intended mutations and diverse in-frame small insertions/deletions were confirmed, indicating that the obtained sublines are useful for molecular pharmacological evaluation of the NT5C2 and PRPS1 gene mutations.
Collapse
Affiliation(s)
- Thao Thu Thi Nguyen
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Yoichi Tanaka
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Masashi Sanada
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Masumi Hosaka
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Minori Tamai
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Keiko Kagami
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Chiaki Komatsu
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Shinpei Somazu
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Daisuke Harama
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Shin Kasai
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Atsushi Watanabe
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Koushi Akahane
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Kumiko Goi
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| | - Takeshi Inukai
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan (T.T.T.N., M.T., K.K., C.K., S.S., D.H., S.K., A.W., K.A., K.G., T.I.); Division of Medicinal Safety Science, National Institutes of Health Sciences, Kanagawa, Japan (Y.T.); and Advanced Diagnostic Research Department, Clinical Research Center, National Hospital Organization, Nagoya Medical Center, Japan (M.S., M.H.)
| |
Collapse
|
10
|
Tamai M, Fujisawa S, Nguyen TTT, Komatsu C, Kagami K, Kamimoto K, Omachi K, Kasai S, Harama D, Watanabe A, Akahane K, Goi K, Naka K, Kaname T, Teshima T, Inukai T. Creation of Philadelphia chromosome by CRISPR/Cas9-mediated double cleavages on BCR and ABL1 genes as a model for initial event in leukemogenesis. Cancer Gene Ther 2023; 30:38-50. [PMID: 35999358 PMCID: PMC9842507 DOI: 10.1038/s41417-022-00522-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/27/2022] [Accepted: 08/04/2022] [Indexed: 01/21/2023]
Abstract
The Philadelphia (Ph) chromosome was the first translocation identified in leukemia. It is supposed to be generated by aberrant ligation between two DNA double-strand breaks (DSBs) at the BCR gene located on chromosome 9q34 and the ABL1 gene located on chromosome 22q11. Thus, mimicking the initiation process of translocation, we induced CRISPR/Cas9-mediated DSBs simultaneously at the breakpoints of the BCR and ABL1 genes in a granulocyte-macrophage colony-stimulating factor (GM-CSF) dependent human leukemia cell line. After transfection of two single guide RNAs (sgRNAs) targeting intron 13 of the BCR gene and intron 1 of the ABL1 gene, a factor-independent subline was obtained. In the subline, p210 BCR::ABL1 and its reciprocal ABL1::BCR fusions were generated as a result of balanced translocation corresponding to the Ph chromosome. Another set of sgRNAs targeting intron 1 of the BCR gene and intron 1 of the ABL1 gene induced a factor-independent subline expressing p190 BCR::ABL1. Both p210 and p190 BCR::ABL1 induced factor-independent growth by constitutively activating intracellular signaling pathways for transcriptional regulation of cell cycle progression and cell survival that are usually regulated by GM-CSF. These observations suggested that simultaneous DSBs at the BCR and ABL1 gene breakpoints are initiation events for oncogenesis in Ph+ leukemia. (200/200 words).
Collapse
Affiliation(s)
- Minori Tamai
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan.
| | - Shinichi Fujisawa
- Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Hokkaido, Japan
| | - Thao T T Nguyen
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Chiaki Komatsu
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Keiko Kagami
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kenji Kamimoto
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St Louis, MO, USA
| | - Kohei Omachi
- Division of Nephrology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Shin Kasai
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Daisuke Harama
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Atsushi Watanabe
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Koshi Akahane
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kumiko Goi
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kazuhito Naka
- Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Tadashi Kaname
- Department of Genome Medicine, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Takanori Teshima
- Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Hokkaido, Japan
| | - Takeshi Inukai
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| |
Collapse
|
11
|
Hiroki H, Akahane K, Inukai T, Morio T, Takagi M. Synergistic effect of combined PI3 kinase inhibitor and PARP inhibitor treatment on BCR/ABL1-positive acute lymphoblastic leukemia cells. Int J Hematol 2022; 117:748-758. [PMID: 36575328 DOI: 10.1007/s12185-022-03520-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022]
Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) function by inhibiting base excision repair and inducing synthetic lethality in homologous recombination repair-deficient cells, such as BRCA1/2-mutated cancer cells. The BCR/ABL1 fusion protein causes dysregulated cell proliferation and is responsible for chronic myelogenous leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ALL). BCR/ABL1 also induces genomic instability by downregulating BRCA1. We investigated the effect of the PARPi, olaparib, against Ph+ALL cell lines and found that they show variable sensitivity, presumably due to cancer-associated genetic alterations other than BCR/ABL1. To investigate the reasons for the variable responses of Ph+ALL cells to PARPi treatment, we analyzed the transcriptomes of olaparib-sensitive and -resistant Ph+ALL cell lines, which revealed that activation of the phosphatidylinositol 3-kinase (PI3K) pathway was a hallmark of PARPi resistance. Based on these findings, we examined the effects of adding a PI3K inhibitor (PI3Ki) to PARPi treatment to overcome PARPi insensitivity in Ph+ALL cell lines. Combination with PI3Ki increased PARPi cytotoxicity in PARPi-resistant Ph+ALL cell lines. Tyrosine kinase inhibitor (TKI) therapy is the gold standard for Ph+ALL, and, based on our findings, we propose that PARPi combined with TKI and PI3K inhibition could be a novel therapeutic strategy for Ph+ALL.
Collapse
Affiliation(s)
- Haruka Hiroki
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Koshi Akahane
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Takeshi Inukai
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Tomohiro Morio
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Masatoshi Takagi
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.
| |
Collapse
|
12
|
Nguyen TTT, Tamai M, Harama D, Kagami K, Kasai S, Watanabe A, Akahane K, Goi K, Inukai T. Introduction of the T315I gatekeeper mutation of BCR/ABL1 into a Philadelphia chromosome-positive lymphoid leukemia cell line using the CRISPR/Cas9 system. Int J Hematol 2022; 116:534-543. [PMID: 35524023 DOI: 10.1007/s12185-022-03369-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 04/20/2022] [Accepted: 04/20/2022] [Indexed: 11/30/2022]
Abstract
Imatinib and second-generation tyrosine kinase inhibitors (TKIs) have dramatically improved the prognosis of Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). However, overcoming TKI resistance due to the T315I gatekeeper mutation of BCR/ABL1 is crucial for further improving the prognosis. The clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system is appropriate for establishing a human model of Ph+ ALL with the T315I mutation, because it can induce specific mutations via homologous recombination (HR) repair in cells with intact endogenous HR pathway. Here we used CRISPR/Cas9 to introduce the T315I mutation into the Ph+ lymphoid leukemia cell line KOPN55bi, which appeared to have an active HR pathway based on its resistance to a poly (ADP-Ribose) polymerase-1 inhibitor. Single-guide RNA targeting at codon 315 and single-strand oligodeoxynucleotide containing ACT to ATT nucleotide transition at codon 315 were electroporated with recombinant Cas9 protein. Dasatinib-resistant sublines were obtained after one-month selection with the therapeutic concentration of dasatinib, leading to T315I mutation acquisition through HR. T315I-acquired sublines were highly resistant to imatinib and second-generation TKIs but moderately sensitive to the therapeutic concentration of ponatinib. This authentic human model is helpful for developing new therapeutic strategies overcoming TKI resistance in Ph+ ALL due to T315I mutation.
Collapse
Affiliation(s)
- Thao T T Nguyen
- Department of Pediatrics, School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Minori Tamai
- Department of Pediatrics, School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Daisuke Harama
- Department of Pediatrics, School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Keiko Kagami
- Department of Pediatrics, School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Shin Kasai
- Department of Pediatrics, School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Atsushi Watanabe
- Department of Pediatrics, School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Koshi Akahane
- Department of Pediatrics, School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Kumiko Goi
- Department of Pediatrics, School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Takeshi Inukai
- Department of Pediatrics, School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan.
| |
Collapse
|
13
|
Genomic Mutations of the STAT5 Transcription Factor Are Associated with Human Cancer and Immune Diseases. Int J Mol Sci 2022; 23:ijms231911297. [PMID: 36232600 PMCID: PMC9569778 DOI: 10.3390/ijms231911297] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022] Open
Abstract
Signal transducer and activation of transcription 5 (STAT5) is a key transcription factor that regulates various biological processes in mammalian development. Aberrant regulation of STAT5 has also been causally linked to many diseases, including cancers and immune-related diseases. Although persistent activation of STAT5 due to dysregulation of the signaling cascade has been reported to be associated with the progression of solid tumors and leukemia, various genomic mutations of STAT5 have also been found to cause a wide range of diseases. The present review comprehensively summarizes results of recent studies evaluating the intrinsic function of STAT5 and the link between STAT5 mutations and human diseases. This review also describes the types of disease models useful for investigating the mechanism underlying STAT5-driven disease progression. These findings provide basic knowledge for understanding the regulatory mechanisms of STAT5 and the progression of various diseases resulting from aberrant regulation of STAT5. Moreover, this review may provide insights needed to create optimal disease models that reflect human disease associated STAT5 mutations and to design gene therapies to correct STAT5 mutations.
Collapse
|
14
|
Nussinov R, Tsai CJ, Jang H. Anticancer drug resistance: An update and perspective. Drug Resist Updat 2021; 59:100796. [PMID: 34953682 PMCID: PMC8810687 DOI: 10.1016/j.drup.2021.100796] [Citation(s) in RCA: 217] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/08/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022]
Abstract
Driver mutations promote initiation and progression of cancer. Pharmacological treatment can inhibit the action of the mutant protein; however, drug resistance almost invariably emerges. Multiple studies revealed that cancer drug resistance is based upon a plethora of distinct mechanisms. Drug resistance mutations can occur in the same protein or in different proteins; as well as in the same pathway or in parallel pathways, bypassing the intercepted signaling. The dilemma that the clinical oncologist is facing is that not all the genomic alterations as well as alterations in the tumor microenvironment that facilitate cancer cell proliferation are known, and neither are the alterations that are likely to promote metastasis. For example, the common KRasG12C driver mutation emerges in different cancers. Most occur in NSCLC, but some occur, albeit to a lower extent, in colorectal cancer and pancreatic ductal carcinoma. The responses to KRasG12C inhibitors are variable and fall into three categories, (i) new point mutations in KRas, or multiple copies of KRAS G12C which lead to higher expression level of the mutant protein; (ii) mutations in genes other than KRAS; (iii) original cancer transitioning to other cancer(s). Resistance to adagrasib, an experimental antitumor agent exerting its cytotoxic effect as a covalent inhibitor of the G12C KRas, indicated that half of the cases present multiple KRas mutations as well as allele amplification. Redundant or parallel pathways included MET amplification; emerging driver mutations in NRAS, BRAF, MAP2K1, and RET; gene fusion events in ALK, RET, BRAF, RAF1, and FGFR3; and loss-of-function mutations in NF1 and PTEN tumor suppressors. In the current review we discuss the molecular mechanisms underlying drug resistance while focusing on those emerging to common targeted cancer drivers. We also address questions of why cancers with a common driver mutation are unlikely to evolve a common drug resistance mechanism, and whether one can predict the likely mechanisms that the tumor cell may develop. These vastly important and tantalizing questions in drug discovery, and broadly in precision medicine, are the focus of our present review. We end with our perspective, which calls for target combinations to be selected and prioritized with the help of the emerging massive compute power which enables artificial intelligence, and the increased gathering of data to overcome its insatiable needs.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, MD, 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
| | - Chung-Jung Tsai
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, MD, 21702, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, MD, 21702, USA
| |
Collapse
|
15
|
Combination of tyrosine kinase inhibitors and the MCL1 inhibitor S63845 exerts synergistic antitumorigenic effects on CML cells. Cell Death Dis 2021; 12:875. [PMID: 34564697 PMCID: PMC8464601 DOI: 10.1038/s41419-021-04154-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 08/22/2021] [Accepted: 09/08/2021] [Indexed: 12/17/2022]
Abstract
Tyrosine kinase inhibitor (TKI) treatment has dramatically improved the survival of chronic myeloid leukemia (CML) patients, but measurable residual disease typically persists. To more effectively eradicate leukemia cells, simultaneous targeting of BCR-ABL1 and additional CML-related survival proteins has been proposed. Notably, several highly specific myeloid cell leukemia 1 (MCL1) inhibitors have recently entered clinical trials for various hematologic malignancies, although not for CML, reflecting the insensitivity of CML cell lines to single MCL1 inhibition. Here, we show that combining TKI (imatinib, nilotinib, dasatinib, or asciminib) treatment with the small-molecule MCL1 inhibitor S63845 exerted strong synergistic antiviability and proapoptotic effects on CML lines and CD34+ stem/progenitor cells isolated from untreated CML patients in chronic phase. Using wild-type BCR-ABL1-harboring CML lines and their T315I-mutated sublines (generated by CRISPR/Cas9-mediated homologous recombination), we prove that the synergistic proapoptotic effect of the drug combination depended on TKI-mediated BCR-ABL1 inhibition, but not on TKI-related off-target mechanisms. Moreover, we demonstrate that colony formation of CML but not normal hematopoietic stem/progenitor cells became markedly reduced upon combination treatment compared to imatinib monotherapy. Our results suggest that dual targeting of MCL1 and BCR-ABL1 activity may efficiently eradicate residual CML cells without affecting normal hematopoietic stem/progenitors.
Collapse
|
16
|
Axitinib in Ponatinib-Resistant B-Cell Acute Lymphoblastic Leukemia Harboring a T315L Mutation. Int J Mol Sci 2020; 21:ijms21249724. [PMID: 33419251 PMCID: PMC7765866 DOI: 10.3390/ijms21249724] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/14/2020] [Accepted: 12/18/2020] [Indexed: 12/20/2022] Open
Abstract
Adult acute lymphoblastic leukemia (ALL) with BCR-ABL1 rearrangement (Philadelphia chromosome, Ph) is a hematological aggressive disease with a fatal outcome in more than 50% of cases. Tyrosine kinase inhibitors (TKIs) targeting the activity of BCR-ABL1 protein have improved the prognosis; however, relapses are frequent because of acquired somatic mutations in the BCR-ABL1 kinase domain causing resistance to first, second and third generation TKIs. Axitinib has shown in vitro and ex vivo activity in blocking ABL1; however, clinical trials exploring its efficacy in ALL are missing. Here, we presented a 77-year-old male with a diagnosis of Ph positive ALL resistant to ponatinib and carrying a rare threonine to leucine (T315L) mutation on BCR-ABL1 gene. The patient was treated with axitinib at 5 mg/twice daily as salvage therapy showing an immediate although transient benefit with an overall survival of 9.3 months. Further dose-finding and randomized clinical trials are required to assess the real efficacy of axitinib for adult Ph positive ALL resistant to third generation TKIs.
Collapse
|
17
|
Giudice V, Vecchione C, Selleri C. Cardiotoxicity of Novel Targeted Hematological Therapies. Life (Basel) 2020; 10:life10120344. [PMID: 33322351 PMCID: PMC7763613 DOI: 10.3390/life10120344] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/16/2022] Open
Abstract
Chemotherapy-related cardiac dysfunction, also known as cardiotoxicity, is a group of drug-related adverse events negatively affecting myocardial structure and functions in patients who received chemotherapy for cancer treatment. Clinical manifestations can vary from life-threatening arrythmias to chronic conditions, such as heart failure or hypertension, which dramatically reduce quality of life of cancer survivors. Standard chemotherapy exerts its toxic effect mainly by inducing oxidative stress and genomic instability, while new targeted therapies work by interfering with signaling pathways important not only in cancer cells but also in myocytes. For example, Bruton’s tyrosine kinase (BTK) inhibitors interfere with class I phosphoinositide 3-kinase isoforms involved in cardiac hypertrophy, contractility, and regulation of various channel forming proteins; thus, off-target effects of BTK inhibitors are associated with increased frequency of arrhythmias, such as atrial fibrillation, compared to standard chemotherapy. In this review, we summarize current knowledge of cardiotoxic effects of targeted therapies used in hematology.
Collapse
Affiliation(s)
- Valentina Giudice
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.V.); (C.S.)
- Clinical Pharmacology, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, 84131 Salerno, Italy
- Correspondence: ; Tel.: +39-089-672-493
| | - Carmine Vecchione
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.V.); (C.S.)
- IRCCS Neuromed (Mediterranean Neurological Institute), 86077 Pozzilli, Italy
| | - Carmine Selleri
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.V.); (C.S.)
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, 84131 Salerno, Italy
| |
Collapse
|
18
|
Chen Y, Zheng J, Gan D, Chen Y, Zhang N, Chen Y, Lin Z, Wang W, Chen H, Lin D, Hu J. E35 ablates acute leukemia stem and progenitor cells in vitro and in vivo. J Cell Physiol 2020; 235:8023-8034. [PMID: 31960417 PMCID: PMC7540425 DOI: 10.1002/jcp.29457] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 01/03/2020] [Indexed: 12/11/2022]
Abstract
Leukemia stem cells (LSCs) have critical functions in acute leukemia (AL) pathogenesis, participating in its initiation and relapse. Thus, identifying new molecules to eradicate LSCs represents a high priority for AL management. This work identified E35, a novel Emodin derivative, which strongly inhibited growth and enhanced apoptosis of AL stem cell lines, and primary stem and progenitor cells from AL cases, while sparing normal hematopoietic cells. Furthermore, functional assays in cultured cells and animals suggested that E35 preferentially ablated primitive leukemia cell populations without impairing their normal counterparts. Moreover, molecular studies showed that E35 remarkably downregulated drug-resistant gene and dramatically inhibited the Akt/mammalian target of rapamycin signaling pathway. Notably, the in vivo anti-LSC activity of E35 was further confirmed in murine xenotransplantation models. Collectively, these findings indicate E35 constitutes a novel therapeutic candidate for AL, potentially targeting leukemia stem and progenitor cells.
Collapse
Affiliation(s)
- Yingyu Chen
- Department of HematologyFujian Institute of HematologyFujian Medical University Union HospitalFuzhouFujianChina
| | - Jing Zheng
- Department of HematologyFujian Institute of HematologyFujian Medical University Union HospitalFuzhouFujianChina
| | - Donghui Gan
- Department of HematologyFujian Institute of HematologyFujian Medical University Union HospitalFuzhouFujianChina
- Department of HematologyThe Affiliated Hospital of Putian UniversityPutianFujianChina
| | - Yanxin Chen
- Department of HematologyFujian Institute of HematologyFujian Medical University Union HospitalFuzhouFujianChina
| | - Na Zhang
- Department of HematologyFujian Institute of HematologyFujian Medical University Union HospitalFuzhouFujianChina
| | - Yuwen Chen
- Department of HematologyFujian Institute of HematologyFujian Medical University Union HospitalFuzhouFujianChina
| | - Zhenxing Lin
- Department of HematologyFujian Institute of HematologyFujian Medical University Union HospitalFuzhouFujianChina
| | - Wenfeng Wang
- Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of ChemistryFuzhou UniversityFuzhouFujianChina
| | - Haijun Chen
- Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of ChemistryFuzhou UniversityFuzhouFujianChina
| | - Donghong Lin
- Department of Clinical LaboratorySchool of Medical Technology and EngineeringFujian Medical UniversityFujianChina
| | - Jianda Hu
- Department of HematologyFujian Institute of HematologyFujian Medical University Union HospitalFuzhouFujianChina
| |
Collapse
|
19
|
Kumar R, Pereira RS, Zanetti C, Minciacchi VR, Merten M, Meister M, Niemann J, Dietz MS, Rüssel N, Schnütgen F, Tamai M, Akahane K, Inukai T, Oellerich T, Kvasnicka HM, Pfeifer H, Nicolini FE, Heilemann M, Van Etten RA, Krause DS. Specific, targetable interactions with the microenvironment influence imatinib-resistant chronic myeloid leukemia. Leukemia 2020; 34:2087-2101. [PMID: 32439895 PMCID: PMC7387317 DOI: 10.1038/s41375-020-0866-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 04/29/2020] [Accepted: 05/11/2020] [Indexed: 12/30/2022]
Abstract
Therapy resistance in leukemia may be due to cancer cell-intrinsic and/or -extrinsic mechanisms. Mutations within BCR-ABL1, the oncogene giving rise to chronic myeloid leukemia (CML), lead to resistance to tyrosine kinase inhibitors (TKI), and some are associated with clinically more aggressive disease and worse outcome. Using the retroviral transduction/transplantation model of CML and human cell lines we faithfully recapitulate accelerated disease course in TKI resistance. We show in various models, that murine and human imatinib-resistant leukemia cells positive for the oncogene BCR-ABL1T315I differ from BCR-ABL1 native (BCR-ABL1) cells with regards to niche location and specific niche interactions. We implicate a pathway via integrin β3, integrin-linked kinase (ILK) and its role in deposition of the extracellular matrix (ECM) protein fibronectin as causative of these differences. We demonstrate a trend towards a reduced BCR-ABL1T315I+ tumor burden and significantly prolonged survival of mice with BCR-ABL1T315I+ CML treated with fibronectin or an ILK inhibitor in xenogeneic and syngeneic murine transplantation models, respectively. These data suggest that interactions with ECM proteins via the integrin β3/ILK-mediated signaling pathway in BCR-ABL1T315I+ cells differentially and specifically influence leukemia progression. Niche targeting via modulation of the ECM may be a feasible therapeutic approach to consider in this setting.
Collapse
Affiliation(s)
- Rahul Kumar
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596, Frankfurt am Main, Germany
| | - Raquel S Pereira
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596, Frankfurt am Main, Germany
| | - Costanza Zanetti
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596, Frankfurt am Main, Germany
| | - Valentina R Minciacchi
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596, Frankfurt am Main, Germany
| | - Maximilian Merten
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596, Frankfurt am Main, Germany
| | - Melanie Meister
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596, Frankfurt am Main, Germany
| | - Julian Niemann
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596, Frankfurt am Main, Germany
| | - Marina S Dietz
- Institute for Physical and Theoretical Chemistry, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Nina Rüssel
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596, Frankfurt am Main, Germany
| | - Frank Schnütgen
- Department of Internal Medicine, Hematology/Oncology, Goethe University, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt am Main, Germany
| | - Minori Tamai
- Department of Pediatrics, School of Medicine, University of Yamanashi, Chuo, Japan
| | - Koshi Akahane
- Department of Pediatrics, School of Medicine, University of Yamanashi, Chuo, Japan
| | - Takeshi Inukai
- Department of Pediatrics, School of Medicine, University of Yamanashi, Chuo, Japan
| | - Thomas Oellerich
- Department of Internal Medicine, Hematology/Oncology, Goethe University, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hans Michael Kvasnicka
- Senckenberg Institute of Pathology, Goethe University Frankfurt, 60590, Frankfurt am Main, Germany
| | - Heike Pfeifer
- Department of Internal Medicine, Hematology/Oncology, Goethe University, Frankfurt am Main, Germany
| | - Franck E Nicolini
- Department of Hematology and INSERM U 1052, CRCL, Centre Léon Bérard, 69373, Lyon Cedex, France
| | - Mike Heilemann
- Institute for Physical and Theoretical Chemistry, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Richard A Van Etten
- Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, 92697, USA
| | - Daniela S Krause
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596, Frankfurt am Main, Germany.
- Department of Internal Medicine, Hematology/Oncology, Goethe University, Frankfurt am Main, Germany.
- Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt am Main, Germany.
- German Cancer Consortium (DKTK), Heidelberg, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|