1
|
Trojani A, Pungolino E, Di Camillo B, Bossi LE, Palumbo C, D’adda M, Perego A, Turrini M, Elena C, Borin LM, Iurlo A, Malato S, Spina F, Latargia ML, Spedini P, Artale S, Anghilieri M, Carraro MC, Bucelli C, Beghini A, Cairoli R. Bone Marrow CD34+/lin- Cells of Patients with Chronic-Phase Chronic Myeloid Leukemia (CP-CML) After 12 Months of Nilotinib Treatment Exhibit a Different Gene Expression Signature Compared to the Diagnosis and the Corresponding Cells from Healthy Subjects. Cancers (Basel) 2025; 17:1022. [PMID: 40149355 PMCID: PMC11940473 DOI: 10.3390/cancers17061022] [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: 02/13/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Chronic-Phase Chronic Myeloid Leukemia (C-PCML) is defined by the presence of the BCR-ABL1 fusion gene, which encodes a tyrosine kinase protein that drives the uncontrolled proliferation and survival of leukemic stem cells (LSCs). Nilotinib, a tyrosine kinase inhibitor, targets the activity of BCR-ABL1 by reducing aberrant signaling pathways, which drive the regeneration of LSCs. Despite nilotinib's action, a population of resilient LSCs persist in the bone marrow (BM) and can indeed drive relapse and progression in CML patients. METHODS Our study investigated the gene expression profiling (GEP) of BM CD34+/lin- cells from 79 CP-CML patients at diagnosis, compared to the BM CD34+/lin- cells from the same patients after 12 months of nilotinib treatment and to the normal counterpart cells from 10 donors (CTRLs). RESULTS GEP analyses identified 3012 significantly differentially expressed genes across these comparisons. Among these, we focused on certain key genes associated with eight crucial KEGG pathways: CML, cell cycle, JAK-STAT, PI3K-Akt, MAPK, Ras, NF-kB, and ABC transporters. Within these pathways, we observed the up-regulation of several genes at diagnosis compared to both 12 months of nilotinib treatment and the CTRLs. CONCLUSIONS We observed that certain transcriptome features present at diagnosis persisted after 12 months of nilotinib treatment, compared to CTRLs. This suggests that nilotinib may exert selective pressure, potentially supporting the survival and self-renewal of LSCs. Future insights into these pathways could help identify therapeutic targets to improve outcomes in CML.
Collapse
Affiliation(s)
- Alessandra Trojani
- Department of Hematology and Oncology, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (E.P.); (L.E.B.); (C.P.); (F.S.); (R.C.)
| | - Ester Pungolino
- Department of Hematology and Oncology, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (E.P.); (L.E.B.); (C.P.); (F.S.); (R.C.)
| | - Barbara Di Camillo
- Department of Information Engineering, University of Padova, 35131 Padova, Italy;
| | - Luca Emanuele Bossi
- Department of Hematology and Oncology, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (E.P.); (L.E.B.); (C.P.); (F.S.); (R.C.)
| | - Cassandra Palumbo
- Department of Hematology and Oncology, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (E.P.); (L.E.B.); (C.P.); (F.S.); (R.C.)
| | - Mariella D’adda
- Department of Hematology and Oncology, ASST Spedali Civili Brescia, 25123 Brescia, Italy;
| | - Alessandra Perego
- Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy; (A.P.); (L.M.B.)
| | - Mauro Turrini
- Department of Internal Medicine, Valduce Hospital, 22100 Como, Italy;
| | - Chiara Elena
- Department of Hematology Oncology, Foundation IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Lorenza Maria Borin
- Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy; (A.P.); (L.M.B.)
| | - Alessandra Iurlo
- Hematology Division, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.I.); (C.B.)
| | - Simona Malato
- Hematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Unit, 20123 Milan, Italy;
| | - Francesco Spina
- Department of Hematology and Oncology, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (E.P.); (L.E.B.); (C.P.); (F.S.); (R.C.)
| | | | | | | | | | | | - Cristina Bucelli
- Hematology Division, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.I.); (C.B.)
| | - Alessandro Beghini
- Department of Health Sciences, University of Milano, 20146 Milan, Italy;
| | - Roberto Cairoli
- Department of Hematology and Oncology, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (E.P.); (L.E.B.); (C.P.); (F.S.); (R.C.)
| |
Collapse
|
2
|
Golestan A, Zareinejad M, Ramezani A. Comprehensive biomarker profiles in hematological malignancies: improving diagnosis, prognosis, and treatment. Biomark Med 2025; 19:223-238. [PMID: 40015744 PMCID: PMC11916375 DOI: 10.1080/17520363.2025.2471745] [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] [Received: 10/14/2024] [Accepted: 02/21/2025] [Indexed: 03/01/2025] Open
Abstract
Hematological malignancies present substantial challenges in clinical practice due to their heterogeneity and complex biological profiles. In these diseases, biomarkers - measurable indicators of biological states - are indispensable for diagnosis, prognosis, and therapeutic decision-making. Emerging biomarkers are significantly improving outcomes in hematological cancers by enhancing early detection, refining prognostic assessments, enabling personalized treatment approaches, and optimizing overall patient management. This progress translates into better clinical outcomes and more effective strategies to treat and manage malignancies. The field of biomarker discovery has developed from basic morphological and cytogenetic markers to advanced molecular techniques, including polymerase chain reaction (PCR) and next-generation sequencing (NGS), which have significantly enhanced diagnostic accuracy and led to the development of targeted therapies. Additionally, the recent advent of technologies like mass spectrometry and single-cell RNA sequencing enables comprehensive molecular profiling and reveals novel biomarkers that were previously undetectable. Our aim in this manuscript is to provide a comprehensive overview of recent and novel immunohematological biomarkers, their diagnostic and therapeutic applications, and the future directions of this field.
Collapse
Affiliation(s)
- Ali Golestan
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Mohammadrasul Zareinejad
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| | - Amin Ramezani
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
| |
Collapse
|
3
|
El Kassem G, Hillmer J, Boettcher M. Evaluation of Cas13d as a tool for genetic interaction mapping. Nat Commun 2025; 16:1631. [PMID: 39952934 PMCID: PMC11828948 DOI: 10.1038/s41467-025-56747-4] [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] [Received: 06/03/2024] [Accepted: 01/28/2025] [Indexed: 02/17/2025] Open
Abstract
Mapping genetic interactions (GIs) is crucial for understanding genetic network complexity. In this study, we investigate the utility of Cas13d, a CRISPR system targeting RNA, for GI mapping and compare it to Cas9 and Cas12a, two DNA nucleases commonly used for GI mapping. We find that Cas13d induces faster target gene perturbation and generates more uniform cell populations with double perturbations than Cas9 or Cas12a. We then encounter Cas13d gRNA-gRNA interference when concatenating gRNAs targeting different genes into one gRNA array, which we overcome by a dual promoter gRNA expression strategy. Moreover, by concatenating three gRNAs targeting the same gene into one array, we are able to maximize the Cas13d-mediated knockdown effects. Combining these strategies enhances proliferation phenotypes while reducing library size and facilitates reproducible quantification of GIs in oncogenic signaling pathways. Our study highlights the potential of Cas13d for GI mapping, promising advancements in understanding therapeutically relevant drug response pathways.
Collapse
Affiliation(s)
- Ghanem El Kassem
- Universitätsmedizin Halle, Martin Luther University Halle-Wittenberg, Halle (Saale), 06120, Halle, Germany
| | - Jasmine Hillmer
- Universitätsmedizin Halle, Martin Luther University Halle-Wittenberg, Halle (Saale), 06120, Halle, Germany
| | - Michael Boettcher
- Universitätsmedizin Halle, Martin Luther University Halle-Wittenberg, Halle (Saale), 06120, Halle, Germany.
| |
Collapse
|
4
|
Vida L, Horváth B, Egyed M, Kajtár B, Alizadeh H. The Many Faces of Philadelphia: A Mature T-Cell Lymphoma with Variant Philadelphia-Translocation and Duplication of the Philadelphia Chromosome. Hematol Rep 2025; 17:1. [PMID: 39846605 PMCID: PMC11755446 DOI: 10.3390/hematolrep17010001] [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: 10/23/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/24/2025] Open
Abstract
Background: T-cell prolymphocytic leukemia (T-PLL) is a rare mature T-cell lymphoma that is usually associated with poor prognosis and short overall survival. Methods: We present a case of a 61-year-old woman presenting with T-PLL and the leukemic cells harboring BCR::ABL1 (BCR-breakpoint cluster region; ABL1-ABL protooncogene 1) fusion transcripts as the result of a variant of t(9;22)(q34;q11) called Philadelphia translocation: t(9;22;18)(q34;q11;q21). Sequencing revealed a rare BCR transcript with an exon 6 breakpoint corresponding to e6a2 transcripts, which has thus far been reported in only 26 cases of leukemias. Results: After 9 months of follow-up, the disease progressed and required treatment. Following alemtuzumab and chemotherapy, a short course of imatinib therapy stabilized the disease for six months, which was followed by progression and the demise of the patient. Conclusions: To the best of our knowledge, this is the first report of a mature T-cell lymphoma with a variant Philadelphia-translocation and a very rare type of BCR::ABL1 transcript. This case highlights the importance of comprehensive genetic testing of malignancies, as abnormal molecular pathways may be uncovered that may be specifically targeted by drugs.
Collapse
Affiliation(s)
- Livia Vida
- Department of Pathology, Medical School, University of Pécs, Szigeti Str. 12, 7624 Pécs, Hungary; (L.V.)
| | - Bálint Horváth
- Department of Pathology, Medical School, University of Pécs, Szigeti Str. 12, 7624 Pécs, Hungary; (L.V.)
| | - Miklós Egyed
- Department of Internal Medicine, Kaposi Mór Teaching Hospital of Somogy County, Tallián Gy. Str. 20-32, 7400 Kaposvár, Hungary
| | - Béla Kajtár
- Department of Pathology, Medical School, University of Pécs, Szigeti Str. 12, 7624 Pécs, Hungary; (L.V.)
| | - Hussain Alizadeh
- Department of 1st Internal Medicine, Medical School, University of Pécs, Ifjúság Str. 13, 7624 Pécs, Hungary
| |
Collapse
|
5
|
Harris TJ, Trader DJ. Exploration of degrons and their ability to mediate targeted protein degradation. RSC Med Chem 2025:d4md00787e. [PMID: 39867589 PMCID: PMC11758578 DOI: 10.1039/d4md00787e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/12/2024] [Indexed: 01/28/2025] Open
Abstract
Degrons are short amino acid sequences that can facilitate the degradation of protein substrates. They can be classified as either ubiquitin-dependent or -independent based on their interactions with the ubiquitin proteasome system (UPS). These amino acid sequences are often found in exposed regions of proteins serving as either a tethering point for an interaction with an E3 ligase or initiating signaling for the direct degradation of the protein. Recent advancements in the protein degradation field have shown the therapeutic potential of both classes of degrons through leveraging their degradative effects to engage specific protein targets. This review explores what targeted protein degradation applications degrons can be used in and how they have inspired new degrader technology to target a wide variety of protein substrates.
Collapse
Affiliation(s)
- Timothy J Harris
- Department of Pharmaceutical Sciences, University of California Irvine California 92617 USA
| | - Darci J Trader
- Department of Pharmaceutical Sciences, University of California Irvine California 92617 USA
- Department of Chemistry, University of California Irvine California 92617 USA
| |
Collapse
|
6
|
Quezada Meza CP, Salizzato V, Calistri E, Basso M, Zavatti M, Marmiroli S, Salvi M, Carter BZ, Donella-Deana A, Borgo C, Ruzzene M. Critical role of protein kinase CK2 in chronic myeloid leukemia cells harboring the T315I BCR::ABL1 mutation. Int J Biol Macromol 2025; 286:138305. [PMID: 39631575 DOI: 10.1016/j.ijbiomac.2024.138305] [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] [Received: 07/26/2024] [Revised: 11/06/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
Chronic myeloid leukemia (CML) is characterized by the fusion protein BCR::ABL1, a constitutively active tyrosine kinase. The frontline treatment, represented by tyrosine kinase inhibitors (TKIs), has dramatically improved the clinical outcomes of patients. However, TKI resistance through various mechanisms has been reported. In particular, the BCR::ABL11 T315I mutation is associated with resistance to first- and second-generation TKIs and poor survival outcomes. For patients harboring this mutation, treatments with third generation TKIs are indicated, which are however accompanied by adverse events. Protein kinase CK2 is implicated in several human diseases. Although its role in CML has already been proven, its essentialness in T315I-mediated TKI resistance has yet to be investigated. Here we show that CK2 contributes to the aberrantly high signaling pathways in T315I-cells, and that its pharmacological or genetic targeting diminishes those signals, induces apoptosis, and reduces the proliferation and clonogenic potential of T315I-cells. The effects of CK2 inhibition are also observed in the presence of bone marrow stromal cells and under hypoxic conditions, and, remarkably, in patient-derived cells. Moreover, CK2 inhibition or genetic ablation of the CK2α catalytic subunit sensitizes T315I-cells towards TKIs. Collectively, our results suggest the potential benefit of inhibiting CK2 in CML characterized by T315I-dependent resistance.
Collapse
MESH Headings
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Casein Kinase II/genetics
- Casein Kinase II/metabolism
- Casein Kinase II/antagonists & inhibitors
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Mutation
- Apoptosis/drug effects
- Apoptosis/genetics
- Protein Kinase Inhibitors/pharmacology
- Cell Proliferation/drug effects
- Drug Resistance, Neoplasm/genetics
- Cell Line, Tumor
- Signal Transduction
Collapse
Affiliation(s)
| | | | | | - Marco Basso
- Pharmacy, Veneto Institute of Oncology IOV IRCCS, Padua, Italy
| | - Manuela Zavatti
- Dept. Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sandra Marmiroli
- Dept. Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Mauro Salvi
- Dept. Biomedical Sciences, University of Padova, Padova, Italy
| | - Bing Z Carter
- Dept. Leukemia, The University of Texas MD Anderson Cancer Center, Houston, USA
| | | | - Christian Borgo
- Dept. Biomedical Sciences, University of Padova, Padova, Italy; Dept. Medicine, University of Padova, Padova, Italy.
| | - Maria Ruzzene
- Dept. Biomedical Sciences, University of Padova, Padova, Italy.
| |
Collapse
|
7
|
Okabe S, Arai Y, Gotoh A. Targeting the insulin-like growth factor-1 receptor to overcome imatinib resistance in chronic myeloid leukemia. Discov Oncol 2024; 15:835. [PMID: 39719486 DOI: 10.1007/s12672-024-01706-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/13/2024] [Indexed: 12/26/2024] Open
Abstract
Patients with chronic myeloid leukemia (CML) frequently develop resistance to tyrosine kinase inhibitors such as imatinib. In this study, we explored the role of the insulin-like growth factor 1 (IGF-1) signaling pathway in CML and imatinib resistance. An analysis of IGF-1 gene expression using public databases revealed elevated levels of insulin-like growth factor-binding proteins in patients with chronic-phase CML. Further research revealed that IGF-1-related genes were upregulated in patients who were unresponsive to imatinib, suggesting that IGF-1 signaling plays a role in the resistance mechanism. Furthermore, we evaluated the efficacy of linsitinib, a selective insulin-like growth factor-1 receptor (IGF-1R) inhibitor, in inhibiting the growth of CML cell lines, including imatinib-resistant cell lines, and observed a notable decrease in cell viability and an increase in cytotoxicity. The combination of imatinib and linsitinib reduced cell viability and increased caspase-3/7 activity in imatinib-resistant cells. Moreover, silencing of IGF-1R by small interfering ribonucleic acid increased the sensitivity of CML cell lines to imatinib, indicating that IGF-1R could be a strategic target for overcoming resistance. These findings highlight the therapeutic potential of linsitinib and that IGF-1R inhibition may improve the treatment outcomes of patients with imatinib-resistant CML.
Collapse
Affiliation(s)
- Seiichi Okabe
- Department of Hematology, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan.
| | - Yuya Arai
- Department of Hematology, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan
| | - Akihiko Gotoh
- Department of Hematology, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku-Ku, Tokyo, 160-0023, Japan
| |
Collapse
|
8
|
Milenkovic J, Stojanovic D, Velickovic S, Djordjevic B, Marjanovic G, Milojkovic M. Low Renalase Levels in Newly Diagnosed CML: Dysregulation Sensitive to Modulation by Tyrosine Kinase Inhibitors. PATHOPHYSIOLOGY 2024; 31:787-796. [PMID: 39728688 DOI: 10.3390/pathophysiology31040053] [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: 09/04/2024] [Revised: 10/23/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
Background: A dysregulated proinflammatory microenvironment is considered one of the reasons why current therapies of chronic myeloid leukemia (CML) with tyrosine kinase inhibitors (TKI) do not secure disease control. Therefore, the development of BCR-ABL1-independent therapies is encouraged. Renalase (RNLS) is a multifunctional protein that exhibits both enzymatic and non-enzymatic cytokine-like properties, along with potent anti-inflammatory and anti-apoptotic effects. It is expressed in various tissues, including tumors. Methods: We investigated the levels of RNLS in the blood of CML patients in the chronic phase, treatment naïve patients, and those in remission under TKI treatment (either imatinib or nilotinib) and compared them to healthy individuals. Results: Renalase concentration was markedly decreased in treatment-naive CML patients compared to other groups (p = 0.000), while lower levels in the TKI group were not statistically significant compared to controls. The levels correlated negatively with the total leukocyte and neutrophil count (p < 0.05), while a positive correlation was present with CRP levels in treatment naïve patients. Conclusions: Dynamic regulation of RNLS expression and activity is coupled with transcription factors NF-κB and STAT3. Interpretation of our results might rely on differential requirements of activated STATs (STAT3/5) during CML clone development and maintenance, including the observation of RNLS rise upon TKI introduction. Overall, our research provides new insights into the field of hematological malignancies. Unlike other malignancies studied, RNLS plasma levels are significantly decreased in CML. In future perspectives, RNLS could potentially serve as a diagnostic, prognostic, or therapeutic option for these patients.
Collapse
Affiliation(s)
- Jelena Milenkovic
- Department of Pathophysiology, University of Nis, Faculty of Medicine, 18000 Nis, Serbia
| | - Dijana Stojanovic
- Department of Pathophysiology, University of Nis, Faculty of Medicine, 18000 Nis, Serbia
| | - Sanja Velickovic
- Clinic of Hematology, Allergology and Clinical Immunology, University Clinical Center in Nis, 18000 Nis, Serbia
| | - Branka Djordjevic
- Department of Biochemistry, University of Nis, Faculty of Medicine, 18000 Nis, Serbia
| | - Goran Marjanovic
- Clinic of Hematology, Allergology and Clinical Immunology, University Clinical Center in Nis, 18000 Nis, Serbia
| | - Maja Milojkovic
- Department of Pathophysiology, University of Nis, Faculty of Medicine, 18000 Nis, Serbia
| |
Collapse
|
9
|
Sanches VHDS, Lobato CC, Silva LB, dos Santos IVF, Barros EDS, Maciel ADA, Ferreira EFB, da Costa KS, Espejo-Román JM, Rosa JMC, Kimani NM, Santos CBR. Rational Approach to New Chemical Entities with Antiproliferative Activity on Ab1 Tyrosine Kinase Encoded by the BCR-ABL Gene: An Hierarchical Biochemoinformatics Analysis. Pharmaceuticals (Basel) 2024; 17:1491. [PMID: 39598402 PMCID: PMC11597596 DOI: 10.3390/ph17111491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Background: This study began with a search in three databases, totaling six libraries (ChemBridge-DIVERSet, ChemBridge-DIVERSet-EXP, Zinc_Drug Database, Zinc_Natural_Stock, Zinc_FDA_BindingDB, Maybridge) with approximately 2.5 million compounds with the aim of selecting potential inhibitors with antiproliferative activity on the chimeric tyrosine kinase encoded by the BCR-ABL gene. Methods: Through hierarchical biochemoinformatics, ADME/Tox analyses, biological activity prediction, molecular docking simulations, synthetic accessibility and theoretical synthetic routes of promising compounds and their lipophilicity and water solubility were realized. Results: Predictions of toxicological and pharmacokinetic properties (ADME/Tox) using the top100/base (600 structures), in comparison with the commercial drug imatinib, showed that only nine exhibited the desired properties. In the prediction of biological activity, the results of the nine selected structures ranged from 13.7% < Pa < 65.8%, showing them to be potential protein kinase inhibitors. In the molecular docking simulations, the promising molecules LMQC01 and LMQC04 showed significant values in molecular targeting (PDB 1IEP-resolution 2.10 Å). LMQC04 presented better binding affinity (∆G = -12.2 kcal mol-1 with a variation of ±3.6 kcal mol-1) in relation to LMQC01. The LMQC01 and LMQC04 molecules were advanced for molecular dynamics (MD) simulation followed by Molecular Mechanics with generalized Born and Surface Area solvation (MM-GBSA); the comparable, low and stable RMSD and ΔE values for the protein and ligand in each complex suggest that the selected compounds form a stable complex with the Abl kinase domain. This stability is a positive indicator that LMQC01 and LMQC04 can potentially inhibit enzyme function. Synthetic accessibility (SA) analysis performed on the AMBIT and SwissADME webservers showed that LMQC01 and LMQC04 can be considered easy to synthesize. Our in silico results show that these molecules could be potent protein kinase inhibitors with potential antiproliferative activity on tyrosine kinase encoded by the BCR-ABL gene. Conclusions: In conclusion, the results suggest that these ligands, particularly LMQC04, may bind strongly to the studied target and may have appropriate ADME/Tox properties in experimental studies. Considering future in vitro or in vivo assays, we elaborated the theoretical synthetic routes of the promising compounds identified in the present study. Based on our in silico findings, the selected ligands show promise for future studies in developing chronic myeloid leukemia treatments.
Collapse
Affiliation(s)
- Vitor H. da S. Sanches
- Biodiversity and Biotechnology Network of the Legal Amazon, Biotechnology Department, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (V.H.d.S.S.); (C.C.L.); (L.B.S.); (I.V.F.d.S.)
- Graduate Program in Medicinal Chemistry and Molecular Modeling, Federal University of Pará, Belém 66075-110, PA, Brazil
- Laboratory of Modeling and Computational Chemistry, Department of Biological and Health Sciences, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (E.d.S.B.); (A.d.A.M.); (E.F.B.F.)
| | - Cleison C. Lobato
- Biodiversity and Biotechnology Network of the Legal Amazon, Biotechnology Department, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (V.H.d.S.S.); (C.C.L.); (L.B.S.); (I.V.F.d.S.)
- Graduate Program in Medicinal Chemistry and Molecular Modeling, Federal University of Pará, Belém 66075-110, PA, Brazil
- Laboratory of Modeling and Computational Chemistry, Department of Biological and Health Sciences, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (E.d.S.B.); (A.d.A.M.); (E.F.B.F.)
| | - Luciane B. Silva
- Biodiversity and Biotechnology Network of the Legal Amazon, Biotechnology Department, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (V.H.d.S.S.); (C.C.L.); (L.B.S.); (I.V.F.d.S.)
- Graduate Program in Medicinal Chemistry and Molecular Modeling, Federal University of Pará, Belém 66075-110, PA, Brazil
- Laboratory of Modeling and Computational Chemistry, Department of Biological and Health Sciences, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (E.d.S.B.); (A.d.A.M.); (E.F.B.F.)
| | - Igor V. F. dos Santos
- Biodiversity and Biotechnology Network of the Legal Amazon, Biotechnology Department, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (V.H.d.S.S.); (C.C.L.); (L.B.S.); (I.V.F.d.S.)
- Laboratory of Modeling and Computational Chemistry, Department of Biological and Health Sciences, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (E.d.S.B.); (A.d.A.M.); (E.F.B.F.)
| | - Elcimar de S. Barros
- Laboratory of Modeling and Computational Chemistry, Department of Biological and Health Sciences, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (E.d.S.B.); (A.d.A.M.); (E.F.B.F.)
- Graduate Program of Pharmaceutical Innovation, Federal University of Amapá, Macapá 68902-280, AP, Brazil
| | - Alexandre de A. Maciel
- Laboratory of Modeling and Computational Chemistry, Department of Biological and Health Sciences, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (E.d.S.B.); (A.d.A.M.); (E.F.B.F.)
- Graduate Program of Pharmaceutical Innovation, Federal University of Amapá, Macapá 68902-280, AP, Brazil
| | - Elenilze F. B. Ferreira
- Laboratory of Modeling and Computational Chemistry, Department of Biological and Health Sciences, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (E.d.S.B.); (A.d.A.M.); (E.F.B.F.)
- Graduate Program of Pharmaceutical Innovation, Federal University of Amapá, Macapá 68902-280, AP, Brazil
- Laboratory of Organic Chemistry and Biochemistry, University of the State of Amapá, Macapá 68900-070, AP, Brazil
| | - Kauê S. da Costa
- Computational Simulation Laboratory, Institute of Biodiversity, Federal University of Western Pará, Vera Paz Street, w/n Salé, Santarém 68040-255, PA, Brazil;
| | - José M. Espejo-Román
- Department of Pharmaceutical and Organic Chemistry, Faculty of Pharmacy, Campus of Cartuja, University of Granada, 18071 Granada, Spain; (J.M.E.-R.); (J.M.C.R.)
| | - Joaquín M. C. Rosa
- Department of Pharmaceutical and Organic Chemistry, Faculty of Pharmacy, Campus of Cartuja, University of Granada, 18071 Granada, Spain; (J.M.E.-R.); (J.M.C.R.)
| | - Njogu M. Kimani
- Natural Product Chemistry and Computational Drug Discovery Laboratory, Embu P.O. Box 6-60100, Kenya;
| | - Cleydson B. R. Santos
- Biodiversity and Biotechnology Network of the Legal Amazon, Biotechnology Department, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (V.H.d.S.S.); (C.C.L.); (L.B.S.); (I.V.F.d.S.)
- Graduate Program in Medicinal Chemistry and Molecular Modeling, Federal University of Pará, Belém 66075-110, PA, Brazil
- Laboratory of Modeling and Computational Chemistry, Department of Biological and Health Sciences, Federal University of Amapá, Macapá 68903-419, AP, Brazil; (E.d.S.B.); (A.d.A.M.); (E.F.B.F.)
- Graduate Program of Pharmaceutical Innovation, Federal University of Amapá, Macapá 68902-280, AP, Brazil
| |
Collapse
|
10
|
Vadeikienė R, Jakštys B, Laukaitienė D, Šatkauskas S, Juozaitytė E, Ugenskienė R. The Role of Mutated Calreticulin in the Pathogenesis of BCR-ABL1-Negative Myeloproliferative Neoplasms. Int J Mol Sci 2024; 25:9873. [PMID: 39337361 PMCID: PMC11432199 DOI: 10.3390/ijms25189873] [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] [Received: 08/15/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Myeloproliferative neoplasms (MPNs) are characterized by increased proliferation of myeloid lineages in the bone marrow. Calreticulin (CALR) 52 bp deletion and CALR 5 bp insertion have been identified in essential thrombocythemia (ET) and primary myelofibrosis (PMF). There is not much data on the crosstalk between mutated CALR and MPN-related signaling pathways, such as JAK/STAT, PI3K/Akt/mTOR, and Hedgehog. Calreticulin, a multifunctional protein, takes part in many cellular processes. Nevertheless, there is little data on how mutated CALR affects the oxidative stress response and oxidative stress-induced DNA damage, apoptosis, and cell cycle progression. We aimed to investigate the role of the CALR 52 bp deletion and 5 bp insertion in the pathogenesis of MPN, including signaling pathway activation and functional analysis in CALR-mutated cells. Our data indicate that the JAK/STAT and PI3K/Akt/mTOR pathways are activated in CALR-mutated cells, and this activation does not necessarily depend on the CALR and MPL interaction. Moreover, it was found that CALR mutations impair calreticulin function, leading to reduced responses to oxidative stress and DNA damage. It was revealed that the accumulation of G2/M-CALR-mutated cells indicates that oxidative stress-induced DNA damage is difficult to repair. Taken together, this study contributes to a deeper understanding of the specific molecular mechanisms underlying CALR-mutated MPNs.
Collapse
Affiliation(s)
- Roberta Vadeikienė
- Oncology Research Laboratory, Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| | - Baltramiejus Jakštys
- Research on Delivery of Medicine and Genes Cluster, Faculty of Natural Sciences, Vytautas Magnus University, LT-44001 Kaunas, Lithuania
| | - Danguolė Laukaitienė
- Oncology Research Laboratory, Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| | - Saulius Šatkauskas
- Research on Delivery of Medicine and Genes Cluster, Faculty of Natural Sciences, Vytautas Magnus University, LT-44001 Kaunas, Lithuania
| | - Elona Juozaitytė
- Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| | - Rasa Ugenskienė
- Oncology Research Laboratory, Institute of Oncology, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
- Department of Genetics and Molecular Medicine, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| |
Collapse
|
11
|
Eshaq AM, Flanagan TW, Hassan SY, Al Asheikh SA, Al-Amoudi WA, Santourlidis S, Hassan SL, Alamodi MO, Bendhack ML, Alamodi MO, Haikel Y, Megahed M, Hassan M. Non-Receptor Tyrosine Kinases: Their Structure and Mechanistic Role in Tumor Progression and Resistance. Cancers (Basel) 2024; 16:2754. [PMID: 39123481 PMCID: PMC11311543 DOI: 10.3390/cancers16152754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/30/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Protein tyrosine kinases (PTKs) function as key molecules in the signaling pathways in addition to their impact as a therapeutic target for the treatment of many human diseases, including cancer. PTKs are characterized by their ability to phosphorylate serine, threonine, or tyrosine residues and can thereby rapidly and reversibly alter the function of their protein substrates in the form of significant changes in protein confirmation and affinity for their interaction with protein partners to drive cellular functions under normal and pathological conditions. PTKs are classified into two groups: one of which represents tyrosine kinases, while the other one includes the members of the serine/threonine kinases. The group of tyrosine kinases is subdivided into subgroups: one of them includes the member of receptor tyrosine kinases (RTKs), while the other subgroup includes the member of non-receptor tyrosine kinases (NRTKs). Both these kinase groups function as an "on" or "off" switch in many cellular functions. NRTKs are enzymes which are overexpressed and activated in many cancer types and regulate variable cellular functions in response to extracellular signaling-dependent mechanisms. NRTK-mediated different cellular functions are regulated by kinase-dependent and kinase-independent mechanisms either in the cytoplasm or in the nucleus. Thus, targeting NRTKs is of great interest to improve the treatment strategy of different tumor types. This review deals with the structure and mechanistic role of NRTKs in tumor progression and resistance and their importance as therapeutic targets in tumor therapy.
Collapse
Affiliation(s)
- Abdulaziz M. Eshaq
- Department of Epidemiology and Biostatistics, Milken Institute School of Public Health, George Washington University, Washington, DC 20052, USA;
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Thomas W. Flanagan
- Department of Pharmacology and Experimental Therapeutics, LSU Health Sciences Center, New Orleans, LA 70112, USA;
| | - Sofie-Yasmin Hassan
- Department of Pharmacy, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany;
| | - Sara A. Al Asheikh
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Waleed A. Al-Amoudi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Simeon Santourlidis
- Institute of Cell Therapeutics and Diagnostics, University Medical Center of Duesseldorf, 40225 Duesseldorf, Germany;
| | - Sarah-Lilly Hassan
- Department of Chemistry, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany;
| | - Maryam O. Alamodi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Marcelo L. Bendhack
- Department of Urology, Red Cross University Hospital, Positivo University, Rua Mauá 1111, Curitiba 80030-200, Brazil;
| | - Mohammed O. Alamodi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Youssef Haikel
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaire, Hôpital Civil, Hôpitaux Universitaire de Strasbourg, 67000 Strasbourg, France
| | - Mossad Megahed
- Clinic of Dermatology, University Hospital of Aachen, 52074 Aachen, Germany;
| | - Mohamed Hassan
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, 67000 Strasbourg, France
- Research Laboratory of Surgery-Oncology, Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
12
|
Khalid M, Suhail M, Faisal A, Poombal F, Muhammad Asad Khan F. Chronic Myeloid Leukemia Unveils Its Dark Side: A Rare Case of Megakaryocytic Blast Crisis. Cureus 2024; 16:e67412. [PMID: 39310611 PMCID: PMC11414841 DOI: 10.7759/cureus.67412] [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: 08/15/2024] [Indexed: 09/25/2024] Open
Abstract
Chronic myeloid leukemia (CML) can progress from a chronic phase (CP) to an accelerated phase (AP) or an acute leukemia-like blastic phase (BP). However, transformation into a megakaryoblastic phase is very rare, and such a progression is clinically significant due to its poor prognosis and resistance to standard tyrosine kinase inhibitors (TKIs). This report discusses a case of CML that progressed to a megakaryoblastic phase and the patient's death within a month despite receiving one cycle of daunorubicin, cytarabine, and TKI chemotherapy. A 39-year-old female with CML (CP) initially achieved hematological remission with nilotinib but later presented with B symptoms and cytopenias indicative of disease progression. A complete diagnostic workup was performed, including blood counts, bone marrow examination, flow cytometry, fluorescence in-situ hybridization (FISH), and cytogenetic testing. Peripheral blood and bone marrow evaluation confirmed blast crisis with 84% medium to large-sized blasts with basophilic cytoplasm and cytoplasmic blebs. The blasts were positive for CD41 and CD61 by immunohistochemistry (IHC). The blasts also expressed CD45 (dim), CD34, CD33, CD117, CD41, and CD61 by flow cytometry. While BCR-ABL1 positivity is typically associated with CML (90-95%), the additional findings point towards a transformation to acute megakaryoblastic leukemia (AMKL or AML-M7). The rare instance of CML's transformation to AMKL highlights the need for megakaryocytic markers in diagnostic panels to ensure accurate diagnosis and timely, tailored therapies for improved outcomes.
Collapse
Affiliation(s)
- Mehreen Khalid
- Hematopathology, Armed Forces Institute of Pathology, Rawalpindi, PAK
| | - Maymoona Suhail
- Hematology, Armed Forces Institute of Pathology, Rawalpindi, PAK
| | - Alizah Faisal
- Hematology, Rawalpindi Medical University, Rawalpindi, PAK
| | - Fnu Poombal
- Pathology, Nishtar Medical University, Multan, PAK
| | | |
Collapse
|
13
|
Oliinyk D, Will A, Schneidmadel FR, Böhme M, Rinke J, Hochhaus A, Ernst T, Hahn N, Geis C, Lubeck M, Raether O, Humphrey SJ, Meier F. µPhos: a scalable and sensitive platform for high-dimensional phosphoproteomics. Mol Syst Biol 2024; 20:972-995. [PMID: 38907068 PMCID: PMC11297287 DOI: 10.1038/s44320-024-00050-9] [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: 08/25/2023] [Revised: 06/06/2024] [Accepted: 06/11/2024] [Indexed: 06/23/2024] Open
Abstract
Mass spectrometry has revolutionized cell signaling research by vastly simplifying the analysis of many thousands of phosphorylation sites in the human proteome. Defining the cellular response to perturbations is crucial for further illuminating the functionality of the phosphoproteome. Here we describe µPhos ('microPhos'), an accessible phosphoproteomics platform that permits phosphopeptide enrichment from 96-well cell culture and small tissue amounts in <8 h total processing time. By greatly minimizing transfer steps and liquid volumes, we demonstrate increased sensitivity, >90% selectivity, and excellent quantitative reproducibility. Employing highly sensitive trapped ion mobility mass spectrometry, we quantify ~17,000 Class I phosphosites in a human cancer cell line using 20 µg starting material, and confidently localize ~6200 phosphosites from 1 µg. This depth covers key signaling pathways, rendering sample-limited applications and perturbation experiments with hundreds of samples viable. We employ µPhos to study drug- and time-dependent response signatures in a leukemia cell line, and by quantifying 30,000 Class I phosphosites in the mouse brain we reveal distinct spatial kinase activities in subregions of the hippocampal formation.
Collapse
Affiliation(s)
- Denys Oliinyk
- Functional Proteomics, Jena University Hospital, 07747, Jena, Germany
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany
| | - Andreas Will
- Functional Proteomics, Jena University Hospital, 07747, Jena, Germany
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany
| | - Felix R Schneidmadel
- Functional Proteomics, Jena University Hospital, 07747, Jena, Germany
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany
| | - Maximilian Böhme
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany
- Klinik für Innere Medizin II, Jena University Hospital, 07747, Jena, Germany
| | - Jenny Rinke
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany
- Klinik für Innere Medizin II, Jena University Hospital, 07747, Jena, Germany
| | - Andreas Hochhaus
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany
- Klinik für Innere Medizin II, Jena University Hospital, 07747, Jena, Germany
| | - Thomas Ernst
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany
- Klinik für Innere Medizin II, Jena University Hospital, 07747, Jena, Germany
| | - Nina Hahn
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, 07747, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, 07747, Jena, Germany
| | - Christian Geis
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, 07747, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, 07747, Jena, Germany
| | - Markus Lubeck
- Bruker Daltonics GmbH & Co. KG, 28359, Bremen, Germany
| | | | - Sean J Humphrey
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, 3052, Victoria, Australia.
| | - Florian Meier
- Functional Proteomics, Jena University Hospital, 07747, Jena, Germany.
- Comprehensive Cancer Center Central Germany, 07747, Jena, Germany.
| |
Collapse
|
14
|
Saber BA, Aygan A, Salihi A. Mutations in Genes Producing Nitric Oxide and Hydrogen Sulfide and Their Connection With Apoptotic Genes in Chronic Myeloid Leukemia. Cureus 2024; 16:e61570. [PMID: 38962618 PMCID: PMC11221202 DOI: 10.7759/cureus.61570] [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: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
Background Despite advances in chronic myeloid leukemia (CML) genetics, the role of nitric oxide (NO) and hydrogen sulfide (H2S) gene mutations and their relationship to apoptotic genes is unclear. Therefore, this study investigated NO- and H2S-producing genes' mutations and their interactions with apoptotic genes using Sanger sequencing and next-generation sequencing (NGS). Methodology A complete blood count (CBC) was carried out to measure the total number of white blood cells, while IL-6 levels were assessed in both control and CML patients using an ELISA technique. Sanger sequencing was used to analyze mutations in the CTH and NOS3 genes, whereas NGS was applied to examine mutations on all chromosomes. Results White blood cell (WBC) and granulocyte counts were significantly higher in CML patients compared to controls (p<0.0001), and monocyte counts were similarly higher (p<0.05). Interleukin-6 (IL-6) levels were significantly elevated in CML patients than controls (p<0.0001), indicating a possible link to CML etiology or progression. Multiple mutations have been identified in both genes, notably in CTH exon 12 and the NOS3 genes VNTR, T786C, and G894T. This study also measured IL-6 concentrations using IL-6 assays, identifying its potential as a CML prognostic diagnostic. WBC counts, granulocyte counts, and mid-range absolute counts, or MID counts, were significantly higher in CML patients than in normal control individuals. NGS identified 1643 somatic and sex chromosomal abnormalities and 439 actively expressed genes in CML patients. The findings imply a genomic landscape beyond the BCR-ABL1 mutation in CML development compared to other databases. Conclusion In conclusion, this study advances the understanding of the genetic characteristics of CML by identifying mutations in the NO- and H2S-producing genes and their complex connections with genes involved in apoptosis. The comprehensive genetic profile obtained by Sanger sequencing and NGS provides possibilities for identifying novel targets for therapy and personalized treatments for CML, therefore contributing to developments in hematological diseases.
Collapse
Affiliation(s)
- Bahaaddin A Saber
- Department of Bioengineering and Sciences, Faculty of Applied Sciences, Kahramanmaraş Sütçü Imam University, Kahramanmaraş, TUR
| | - Ashabil Aygan
- Department of Biology, Faculty of Applied Sciences, Kahramanmaraş Sütçü Imam University, Kahramanmaraş, TUR
| | - Abbas Salihi
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, IRQ
| |
Collapse
|
15
|
Gomez EW, De Paula LB, Weimer RD, Hellwig AHDS, Rodrigues GM, Alegretti AP, de Oliveira JR. The potential of circHIPK3 as a biomarker in chronic myeloid leukemia. Front Oncol 2024; 14:1330592. [PMID: 38505596 PMCID: PMC10948418 DOI: 10.3389/fonc.2024.1330592] [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] [Received: 10/31/2023] [Accepted: 02/14/2024] [Indexed: 03/21/2024] Open
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative disorder characterized by leukocytosis and left shift. The primary molecular alteration is the BCR::ABL1, chimeric oncoprotein with tyrosine kinase activity, responsible for the initial oncogenesis of the disease. Therapy of CML was revolutionized with the advent of tyrosine kinase inhibitors, but it is still not considered curative and may present resistance and serious adverse effects. Discoveries in CML inaugurated a new era in cancer treatment and despite all the advances, a new biomarker is needed to detect resistance and adverse effects. Circular RNAs (circRNAs) are a special type of non-coding RNA formed through a process called backsplicing. The majority of circRNAs are derived from protein-coding genes. CircHIPK3 is formed from the second exon of the HIPK3 gene and has been found in various pathologies, including different types of cancer. New approaches have demonstrated the potential of circular RNAs in cancer research, and circHIPK3 has shown promising results. It is often associated with cellular regulatory pathways, suggesting an important role in the molecular dynamics of tumors. The identification of biomarkers is an important tool for therapeutic improvement; thus we review the role of circHIPK3 and its potential as a biomarker in CML.
Collapse
Affiliation(s)
- Eduardo Wandame Gomez
- Laboratory of Cell Biophysics and Inflammation, School of Health and Life Sciences, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratory of Molecular Biology, Laboratory Diagnostic Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Laura Berti De Paula
- Laboratory of Cell Biophysics and Inflammation, School of Health and Life Sciences, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Rafael Diogo Weimer
- Laboratory of Cell Biophysics and Inflammation, School of Health and Life Sciences, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Grazielle Motta Rodrigues
- Laboratory of Molecular Biology, Laboratory Diagnostic Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Ana Paula Alegretti
- Laboratory of Molecular Biology, Laboratory Diagnostic Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Jarbas Rodrigues de Oliveira
- Laboratory of Cell Biophysics and Inflammation, School of Health and Life Sciences, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
16
|
Jabbour E, Apperley J, Cortes J, Rea D, Deininger M, Abruzzese E, Chuah C, DeAngelo DJ, Hochhaus A, Lipton JH, Mauro M, Nicolini F, Pinilla-Ibarz J, Rosti G, Rousselot P, Shah NP, Talpaz M, Vorog A, Ren X, Kantarjian H. Dose modification dynamics of ponatinib in patients with chronic-phase chronic myeloid leukemia (CP-CML) from the PACE and OPTIC trials. Leukemia 2024; 38:475-481. [PMID: 38287132 PMCID: PMC10912029 DOI: 10.1038/s41375-024-02159-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/22/2023] [Accepted: 01/15/2024] [Indexed: 01/31/2024]
Abstract
Ponatinib, the only approved all known-BCR::ABL1 inhibitor, is a third-generation tyrosine-kinase inhibitor (TKI) designed to inhibit BCR::ABL1 with or without any single resistance mutation, including T315I, and induced robust and durable responses at 45 mg/day in patients with CP-CML resistant to second-generation TKIs in the PACE trial. However, cardiovascular toxicities, including arterial occlusive events (AOEs), have emerged as treatment-related AEs within this class of TKIs. The OPTIC trial evaluated the efficacy and safety of ponatinib using a novel, response-based, dose-reduction strategy in patients with CP-CML whose disease is resistant to ≥2 TKIs or who harbor T315I. To assess the dose-response relationship and the effect on the safety of ponatinib, we examined the outcomes of patients with CP-CML enrolled in PACE and OPTIC who received 45 mg/day of ponatinib. A propensity score analysis was used to evaluate AOEs across both trials. Survival rates and median time to achieve ≤1% BCR::ABL1IS in OPTIC were similar or better than in PACE. The outcomes of patients with T315I mutations were robust in both trials. Patients in OPTIC had a lower exposure-adjusted incidence of AOEs compared with those in PACE. This analysis demonstrates that response-based dosing for ponatinib improves treatment tolerance and mitigates cardiovascular risk.
Collapse
MESH Headings
- Humans
- Drug Resistance, Neoplasm
- Leukemia, Myeloid, Chronic-Phase/drug therapy
- Leukemia, Myeloid, Chronic-Phase/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Imidazoles/therapeutic use
- Imidazoles/pharmacology
- Pyridazines/therapeutic use
- Pyridazines/pharmacology
- Fusion Proteins, bcr-abl/genetics
- Protein Kinase Inhibitors/pharmacology
- Antineoplastic Agents/therapeutic use
- Antineoplastic Agents/pharmacology
Collapse
Affiliation(s)
- Elias Jabbour
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | | | | | | | - Michael Deininger
- Versiti Blood Research Institute, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Charles Chuah
- Singapore General Hospital, National Cancer Centre Singapore, Duke-NUS Medical School, Singapore, Singapore
| | | | | | | | | | | | | | | | - Philippe Rousselot
- Hospital Mignot University de Versailles Saint-Quentin-en-Yvelines, Paris, France
| | - Neil P Shah
- University of California San Francisco, San Francisco, CA, USA
| | - Moshe Talpaz
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Alexander Vorog
- Takeda Development Center Americas, Inc., Lexington, MA, USA
| | - Xiaowei Ren
- Takeda Development Center Americas, Inc., Lexington, MA, USA
| | - Hagop Kantarjian
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
17
|
El-Tanani M, Nsairat H, Matalka II, Lee YF, Rizzo M, Aljabali AA, Mishra V, Mishra Y, Hromić-Jahjefendić A, Tambuwala MM. The impact of the BCR-ABL oncogene in the pathology and treatment of chronic myeloid leukemia. Pathol Res Pract 2024; 254:155161. [PMID: 38280275 DOI: 10.1016/j.prp.2024.155161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 01/29/2024]
Abstract
Chronic Myeloid Leukemia (CML) is characterized by chromosomal aberrations involving the fusion of the BCR and ABL genes on chromosome 22, resulting from a reciprocal translocation between chromosomes 9 and 22. This fusion gives rise to the oncogenic BCR-ABL, an aberrant tyrosine kinase identified as Abl protein. The Abl protein intricately regulates the cell cycle by phosphorylating protein tyrosine residues through diverse signaling pathways. In CML, the BCR-ABL fusion protein disrupts the first exon of Abl, leading to sustained activation of tyrosine kinase and resistance to deactivation mechanisms. Pharmacological interventions, such as imatinib, effectively target BCR-ABL's tyrosine kinase activity by binding near the active site, disrupting ATP binding, and inhibiting downstream protein phosphorylation. Nevertheless, the emergence of resistance, often attributed to cap structure mutations, poses a challenge to imatinib efficacy. Current research endeavours are directed towards overcoming resistance and investigating innovative therapeutic strategies. This article offers a comprehensive analysis of the structural attributes of BCR-ABL, emphasizing its pivotal role as a biomarker and therapeutic target in CML. It underscores the imperative for ongoing research to refine treatment modalities and enhance overall outcomes in managing CML.
Collapse
MESH Headings
- Humans
- Imatinib Mesylate/therapeutic use
- Imatinib Mesylate/pharmacology
- Genes, abl
- Pyrimidines/therapeutic use
- Piperazines/therapeutic use
- Benzamides/pharmacology
- Benzamides/therapeutic use
- Drug Resistance, Neoplasm/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Fusion Proteins, bcr-abl/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Protein Kinase Inhibitors/pharmacology
Collapse
Affiliation(s)
- Mohamed El-Tanani
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates; Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan.
| | - Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Ismail I Matalka
- Ras Al Khaimah Medical and Health Sciences University, United Arab Emirates; Department of Pathology and Microbiology, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Yin Fai Lee
- Neuroscience, Psychology & Behaviour, College of Life Sciences, University of Leicester, Leicester LE1 9HN, UK; School of Life Sciences, Faculty of Science and Engineering, Anglia Ruskin University, Cambridge CB1 1PT, UK
| | - Manfredi Rizzo
- Department of Health Promotion, Mother and Childcare, Internal Medicine and Medical Specialties, School of Medicine, University of Palermo, Palermo, Italy
| | - Alaa A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid 21163, Jordan
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Yachana Mishra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, Sarajevo 71000, Bosnia and Herzegovina
| | - Murtaza M Tambuwala
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates; Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, UK.
| |
Collapse
|
18
|
Bochicchio MT, Marconi G, Baldazzi C, Bandini L, Ruggieri F, Lucchesi A, Agostinelli C, Sabattini E, Orsatti A, Ferrari A, Capirossi G, Servili C, Ghelli Luserna di Rorà A, Martinelli G, Simonetti G, Rosti G. ETV6::ABL1-Positive Myeloid Neoplasm: A Case of a Durable Response to Imatinib Mesylate without Additional or Previous Treatment. Int J Mol Sci 2023; 25:118. [PMID: 38203288 PMCID: PMC10779409 DOI: 10.3390/ijms25010118] [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: 10/27/2023] [Revised: 12/08/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
ETV6::ABL1 rearranged neoplasms are rare hematological diseases. To date, about 80 cases have been reported, including myeloid and lymphoid leukemias. The ETV6 gene codes for an ETS family transcription factor and several fusion partners have been described. When translocated, ETV6 causes the constitutive activation of the partner genes. Here, we report the case of a 54-year-old woman with a cryptic insertion of the 3' region of ABL1 in the ETV6 gene. The patient was first diagnosed with idiopathic hypereosinophilic syndrome, according to the clinical history, conventional cytogenetics, standard molecular analyses and pathologist description. Next generation sequencing of diagnosis samples unexpectedly detected both ETV6::ABL1 type A and B fusion transcripts, which were then confirmed by FISH. The diagnosis was Myeloid/Lymphoid neoplasm with ETV6::ABL1 fusion, and the patient received imatinib mesylate treatment. In a follow-up after more than one year, the patient still maintained the molecular and complete hematological responses. This case highlights the importance of timely and proper diagnostics and prompt tyrosine kinase inhibitor treatment.
Collapse
Affiliation(s)
- Maria Teresa Bochicchio
- Biosciences Laboratory, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, FC, Italy; (F.R.); (A.F.); (G.C.); (C.S.); (G.S.)
| | - Giovanni Marconi
- Hematology Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, FC, Italy; (G.M.); (A.L.)
| | - Carmen Baldazzi
- Istituto di Ematologia “Seràgnoli”, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, BO, Italy; (C.B.); (L.B.)
| | - Lorenza Bandini
- Istituto di Ematologia “Seràgnoli”, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, BO, Italy; (C.B.); (L.B.)
| | - Francesca Ruggieri
- Biosciences Laboratory, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, FC, Italy; (F.R.); (A.F.); (G.C.); (C.S.); (G.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40100 Bologna, BO, Italy;
| | - Alessandro Lucchesi
- Hematology Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, FC, Italy; (G.M.); (A.L.)
| | - Claudio Agostinelli
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40100 Bologna, BO, Italy;
- Haematopathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, BO, Italy; (E.S.); (A.O.)
| | - Elena Sabattini
- Haematopathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, BO, Italy; (E.S.); (A.O.)
| | - Agnese Orsatti
- Haematopathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, BO, Italy; (E.S.); (A.O.)
| | - Anna Ferrari
- Biosciences Laboratory, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, FC, Italy; (F.R.); (A.F.); (G.C.); (C.S.); (G.S.)
| | - Giorgia Capirossi
- Biosciences Laboratory, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, FC, Italy; (F.R.); (A.F.); (G.C.); (C.S.); (G.S.)
| | - Chiara Servili
- Biosciences Laboratory, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, FC, Italy; (F.R.); (A.F.); (G.C.); (C.S.); (G.S.)
| | | | - Giovanni Martinelli
- Scientific Directorate, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, FC, Italy;
| | - Giorgia Simonetti
- Biosciences Laboratory, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, FC, Italy; (F.R.); (A.F.); (G.C.); (C.S.); (G.S.)
| | - Gianantonio Rosti
- Hematology Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, FC, Italy; (G.M.); (A.L.)
| |
Collapse
|
19
|
Shahraki S, Bahraini F, Mesbahzadeh B, Sayadi M, Sajjadi SM. Glucose increases proliferation and chemoresistance in chronic myeloid leukemia via decreasing antioxidant Properties of ω-3 polyunsaturated fatty acids in the presence of Iron. Mol Biol Rep 2023; 50:10315-10324. [PMID: 37971569 DOI: 10.1007/s11033-023-08891-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 10/04/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND There is a strong association between hyperglycemia, oxidative stress, inflammation and the onset and progression of diabetes which causes a higher risk of cancer. This study investigated, the effect of concomitant use of omega-3 polyunsaturated fatty acids (ω-3 PUFAs) with iron supplements in hyper-glucose conditions on the K-562 cell line. METHODS The effects of iron, ω-3 PUFAs, and a combination of both on K-562 cells were investigated under normal and high glucose conditions. The impact of these treatments was evaluated using multiple methodologies, including the MTT assay for cell viability, quantification of oxidative stress markers [total antioxidant capacity (TAC) and malondialdehyde (MDA)], and analysis of the cell cycle. Furthermore, the expression levels of TNFα and p53 mRNA were measured using Real-time PCR. RESULTS The co-treatment of ω-3 PUFAs and iron in the presence of high glucose had notable effects, as evidenced by an increase in cell survival, resistance to imatinib chemotherapy, TNFαmRNA expression levels, MDA levels, and percentage of cells in the G2/S phase. Additionally, there was a decrease in the mRNA expression of p53 and TAC levels compared to treatment in the normal-glucose condition. CONCLUSION Hyperglycemic conditions in conjunction with the combined treatment of theω-3 PUFAs and iron, led to reduced anticancer capacity, chemosensitivity, anti-inflammatory and antioxidant properties of the K-562 cells. These effects were found to be mediated by oxidative stress.
Collapse
Affiliation(s)
- Samira Shahraki
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Fatemeh Bahraini
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Behzad Mesbahzadeh
- Department of Physiology, School of Allied Medical Sciences, Birjand University of Medical Sciences, Birjand, Iran
| | - Mahtab Sayadi
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| | - Seyed Mehdi Sajjadi
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|
20
|
Malarz K, Mularski J, Pacholczyk M, Musiol R. Styrylquinazoline derivatives as ABL inhibitors selective for different DFG orientations. J Enzyme Inhib Med Chem 2023; 38:2201410. [PMID: 37070569 PMCID: PMC10120462 DOI: 10.1080/14756366.2023.2201410] [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] [Received: 01/21/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/19/2023] Open
Abstract
Among tyrosine kinase inhibitors, quinazoline-based compounds represent a large and well-known group of multi-target agents. Our previous studies have shown interesting kinases inhibition activity for a series of 4-aminostyrylquinazolines based on the CP-31398 scaffold. Here, we synthesised a new series of styrylquinazolines with a thioaryl moiety in the C4 position and evaluated in detail their biological activity. Our results showed high inhibition potential against non-receptor tyrosine kinases for several compounds. Molecular docking studies showed differential binding to the DFG conformational states of ABL kinase for two derivatives. The compounds showed sub-micromolar activity against leukaemia. Finally, in-depth cellular studies revealed the full landscape of the mechanism of action of the most active compounds. We conclude that S4-substituted styrylquinazolines can be considered as a promising scaffold for the development of multi-kinase inhibitors targeting a desired binding mode to kinases as effective anticancer drugs.
Collapse
Affiliation(s)
- Katarzyna Malarz
- Institute of Physics, University of Silesia in Katowice, Chorzów, Poland
| | - Jacek Mularski
- Institute of Chemistry, University of Silesia in Katowice, Chorzów, Poland
| | - Marcin Pacholczyk
- Department of Systems Biology and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Robert Musiol
- Institute of Chemistry, University of Silesia in Katowice, Chorzów, Poland
| |
Collapse
|
21
|
Lahlil R, Aries A, Scrofani M, Zanetti C, Hennequin D, Drénou B. Stem Cell Responsiveness to Imatinib in Chronic Myeloid Leukemia. Int J Mol Sci 2023; 24:16671. [PMID: 38068992 PMCID: PMC10706348 DOI: 10.3390/ijms242316671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/17/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Chronic myeloid leukemia (CML) is a clonal myeloproliferative disease characterized by the presence of the BCR-ABL fusion gene, which results from the Philadelphia chromosome. Since the introduction of tyrosine kinase inhibitors (TKI) such as imatinib mesylate (IM), the clinical outcomes for patients with CML have improved significantly. However, IM resistance remains the major clinical challenge for many patients, underlining the need to develop new drugs for the treatment of CML. The basis of CML cell resistance to this drug is unclear, but the appearance of additional genetic alterations in leukemic stem cells (LSCs) is the most common cause of patient relapse. However, several groups have identified a rare subpopulation of CD34+ stem cells in adult patients that is present mainly in the bone marrow and is more immature and pluripotent; these cells are also known as very small embryonic-like stem cells (VSELs). The uncontrolled proliferation and a compromised differentiation possibly initiate their transformation to leukemic VSELs (LVSELs). Their nature and possible involvement in carcinogenesis suggest that they cannot be completely eradicated with IM treatment. In this study, we demonstrated that cells from CML patients with the VSELs phenotype (LVSELs) similarly harbor the fusion protein BCR-ABL and are less sensitive to apoptosis than leukemic HSCs after IM treatment. Thus, IM induces apoptosis and reduces the proliferation and mRNA expression of Ki67 more efficiently in LHSCs than in leukemic LVSELs. Finally, we found that the expression levels of some miRNAs are affected in LVSELs. In addition to the tumor suppressor miR-451, both miR-126 and miR-21, known to be responsible for LSC leukemia-initiating capacity, quiescence, and growth, appear to be involved in IM insensitivity of LVSELs CML cell population. Targeting IM-resistant CML leukemic stem cells by acting via the miRNA pathways may represent a promising therapeutic option.
Collapse
MESH Headings
- Adult
- Humans
- Imatinib Mesylate/pharmacology
- Imatinib Mesylate/therapeutic use
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Protein Kinase Inhibitors/metabolism
- Drug Resistance, Neoplasm/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- MicroRNAs/metabolism
- Apoptosis
- Stem Cells/metabolism
- Neoplastic Stem Cells/metabolism
Collapse
Affiliation(s)
- Rachid Lahlil
- Institut de Recherche en Hématologie et Transplantation (IRHT), Hôpital du Hasenrain, 87 Avenue d’Altkirch, 68100 Mulhouse, France; (A.A.); (B.D.)
| | - Anne Aries
- Institut de Recherche en Hématologie et Transplantation (IRHT), Hôpital du Hasenrain, 87 Avenue d’Altkirch, 68100 Mulhouse, France; (A.A.); (B.D.)
| | - Maurice Scrofani
- Institut de Recherche en Hématologie et Transplantation (IRHT), Hôpital du Hasenrain, 87 Avenue d’Altkirch, 68100 Mulhouse, France; (A.A.); (B.D.)
| | - Céline Zanetti
- Institut de Recherche en Hématologie et Transplantation (IRHT), Hôpital du Hasenrain, 87 Avenue d’Altkirch, 68100 Mulhouse, France; (A.A.); (B.D.)
| | - Desline Hennequin
- Institut de Recherche en Hématologie et Transplantation (IRHT), Hôpital du Hasenrain, 87 Avenue d’Altkirch, 68100 Mulhouse, France; (A.A.); (B.D.)
| | - Bernard Drénou
- Institut de Recherche en Hématologie et Transplantation (IRHT), Hôpital du Hasenrain, 87 Avenue d’Altkirch, 68100 Mulhouse, France; (A.A.); (B.D.)
- Laboratoire d’Hématologie, Groupe Hospitalier de la Région de Mulhouse Sud-Alsace, Hôpital E. Muller, 20 Avenue de Dr. Laennec, 68100 Mulhouse, France
| |
Collapse
|
22
|
Solís-Hernández MDJ, Palomares-Báez JP, Herrera-Bucio R, Chacón-García L, Navarro-Santos P. Derivates of 1,6-dihyadroazaazulenes as inhibitors of tyrosine kinases BCR-ABL1 wild type and mutant T315I: a molecular dynamics approach. J Biomol Struct Dyn 2023; 42:13864-13875. [PMID: 37937766 DOI: 10.1080/07391102.2023.2279274] [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] [Received: 07/04/2023] [Accepted: 10/30/2023] [Indexed: 11/09/2023]
Abstract
The protein tyrosine kinase (PTK) produced by the BCR-ABL1 gene has generated significant interest in the development of inhibitors since the presence of punctual mutations causes resistance to currently approved drugs, mainly the T315I mutation has been the most difficult to address. In this work, derivatives of 1,6-dihydroazaazulenes are studied as possible inhibitors of this PTK in its wild form and the mutant T315I. The recognition of the ligands was explored through molecular docking, and the stability of the complexes and their evolution over time was studied using molecular dynamics (MD) simulations. Our results show that complexes are energetically stable and reside on the ATP binding site in all cases during the MD experiments. Interestingly, a few of our proposed ligands presented greater affinity for T315I, finding more favorable binding free energies (ΔG) than the reference drug axitinib. Furthermore, they may act as inhibitors for both isoforms. Our findings are promising because mutation of T315I does not prevent ligand recognition, as detailed in this work, which is very important to conduct further experimental research.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Manuel de Jesus Solís-Hernández
- Instituto de Investigaciones Quimico Biologicas, Universidad Michoacana de San Nicolas de Hidalgo Edificio B-1, Ciudad Universitaria, Michoacán, Mexico
| | | | - Rafael Herrera-Bucio
- Instituto de Investigaciones Quimico Biologicas, Universidad Michoacana de San Nicolas de Hidalgo Edificio B-1, Ciudad Universitaria, Michoacán, Mexico
| | - Luis Chacón-García
- Instituto de Investigaciones Quimico Biologicas, Universidad Michoacana de San Nicolas de Hidalgo Edificio B-1, Ciudad Universitaria, Michoacán, Mexico
| | - Pedro Navarro-Santos
- Instituto de Investigaciones Quimico Biologicas, Universidad Michoacana de San Nicolas de Hidalgo Edificio B-1, Ciudad Universitaria, Michoacán, Mexico
- CONACYT-Universidad Michoacana de San Nicolas de Hidalgo Edificio B-1, Ciudad Universitaria, Michoacán, Mexico
| |
Collapse
|
23
|
Dong H, Chang CD, Gao F, Zhang N, Yan XJ, Wu X, Wang YH. The anti-leukemia activity and mechanisms of shikonin: a mini review. Front Pharmacol 2023; 14:1271252. [PMID: 38026987 PMCID: PMC10651754 DOI: 10.3389/fphar.2023.1271252] [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] [Received: 08/02/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Leukemia encompasses a group of highly heterogeneous diseases that pose a serious threat to human health. The long-term outcome of patients with leukemia still needs to be improved and new effective therapeutic strategies continue to be an unmet clinical need. Shikonin (SHK) is a naphthoquinone derivative that shows multiple biological function includes anti-tumor, anti-inflammatory, and anti-allergic effects. Numerous studies have reported the anti-leukemia activity of SHK during the last 3 decades and there are studies showing that SHK is particularly effective towards various leukemia cells compared to solid tumors. In this review, we will discuss the anti-leukemia effect of SHK and summarize the underlying mechanisms. Therefore, SHK may be a promising agent to be developed as an anti-leukemia drug.
Collapse
Affiliation(s)
- Han Dong
- Department of Geriatrics, Jilin Geriatrics Clinical Research Center, The First Hospital of Jilin University, Changchun, China
| | - Chun-Di Chang
- Department of Neurology, Jilin Province People’s Hospital, Changchun, China
| | - Fei Gao
- Endocrine Department, Qian Wei Hospital of Jilin Province, Changchun, China
| | - Na Zhang
- Electrodiagnosis Department, Jilin Province FAW General Hospital, Changchun, China
| | - Xing-Jian Yan
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Xue Wu
- Department of Geriatrics, Jilin Geriatrics Clinical Research Center, The First Hospital of Jilin University, Changchun, China
| | - Yue-Hui Wang
- Department of Geriatrics, Jilin Geriatrics Clinical Research Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
24
|
Verhagen NE, Koenderink JB, Blijlevens NMA, Janssen JJWM, Russel FGM. Transporter-Mediated Cellular Distribution of Tyrosine Kinase Inhibitors as a Potential Resistance Mechanism in Chronic Myeloid Leukemia. Pharmaceutics 2023; 15:2535. [PMID: 38004514 PMCID: PMC10675650 DOI: 10.3390/pharmaceutics15112535] [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: 10/03/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 11/26/2023] Open
Abstract
Chronic myeloid leukemia (CML) is a hematologic neoplasm characterized by the expression of the BCR::ABL1 oncoprotein, a constitutively active tyrosine kinase, resulting in uncontrolled growth and proliferation of cells in the myeloid lineage. Targeted therapy using tyrosine kinase inhibitors (TKIs) such as imatinib, nilotinib, dasatinib, bosutinib, ponatinib and asciminib has drastically improved the life expectancy of CML patients. However, treatment resistance occurs in 10-20% of CML patients, which is a multifactorial problem that is only partially clarified by the presence of TKI inactivating BCR::ABL1 mutations. It may also be a consequence of a reduction in cytosolic TKI concentrations in the target cells due to transporter-mediated cellular distribution. This review focuses on drug-transporting proteins in stem cells and progenitor cells involved in the distribution of TKIs approved for the treatment of CML. Special attention will be given to ATP-binding cassette transporters expressed in lysosomes, which may facilitate the extracytosolic sequestration of these compounds.
Collapse
Affiliation(s)
- Noor E. Verhagen
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (N.E.V.); (J.B.K.)
| | - Jan B. Koenderink
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (N.E.V.); (J.B.K.)
| | - Nicole M. A. Blijlevens
- Department of Haematology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (N.M.A.B.); (J.J.W.M.J.)
| | - Jeroen J. W. M. Janssen
- Department of Haematology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (N.M.A.B.); (J.J.W.M.J.)
| | - Frans G. M. Russel
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (N.E.V.); (J.B.K.)
| |
Collapse
|
25
|
Zhang J, Ma C, Yu Y, Liu C, Fang L, Rao H. Single amino acid-based PROTACs trigger degradation of the oncogenic kinase BCR-ABL in chronic myeloid leukemia (CML). J Biol Chem 2023; 299:104994. [PMID: 37392851 PMCID: PMC10388202 DOI: 10.1016/j.jbc.2023.104994] [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] [Received: 04/22/2023] [Revised: 06/09/2023] [Accepted: 06/19/2023] [Indexed: 07/03/2023] Open
Abstract
Proteolysis-targeting chimera (PROTAC) that specifically targets harmful proteins for destruction by hijacking the ubiquitin-proteasome system is emerging as a potent anticancer strategy. How to efficiently modulate the target degradation remains a challenging issue. In this study, we employ a single amino acid-based PROTAC, which uses the shortest degradation signal sequence as the ligand of the N-end rule E3 ubiquitin ligases to degrade the fusion protein BCR (breakpoint cluster region)-ABL (Abelson proto-oncogene), an oncogenic kinase that drives the progression of chronic myeloid leukemia. We find that the reduction level of BCR-ABL can be easily adjusted by substituting different amino acids. Furthermore, a single PEG linker is found to achieve the best proteolytic effect. Our efforts have resulted in effective degradation of BCR-ABL protein by the N-end rule pathway and efficient growth inhibition of K562 cells expressing BCR-ABL in vitro and blunted tumor growth in a K562 xenograft tumor model in vivo. The PROTAC presented has unique advantages including lower effective concentration, smaller molecular size, and modular degradation rate. Demonstrating the efficacy of the N-end rule-based PROTACs in vitro and in vivo, our study further expands the limited degradation pathways currently available for PROTACs in vivo and is easily adapted for broader applications in targeted protein degradation.
Collapse
MESH Headings
- Humans
- Proteolysis Targeting Chimera
- Amino Acids
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- K562 Cells
- Ubiquitins
Collapse
Affiliation(s)
- Jianchao Zhang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Caibing Ma
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Yongjun Yu
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Chaowei Liu
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Lijing Fang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China.
| | - Hai Rao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, China; Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
26
|
Salokas K, Dashi G, Varjosalo M. Decoding Oncofusions: Unveiling Mechanisms, Clinical Impact, and Prospects for Personalized Cancer Therapies. Cancers (Basel) 2023; 15:3678. [PMID: 37509339 PMCID: PMC10377698 DOI: 10.3390/cancers15143678] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Cancer-associated gene fusions, also known as oncofusions, have emerged as influential drivers of oncogenesis across a diverse range of cancer types. These genetic events occur via chromosomal translocations, deletions, and inversions, leading to the fusion of previously separate genes. Due to the drastic nature of these mutations, they often result in profound alterations of cellular behavior. The identification of oncofusions has revolutionized cancer research, with advancements in sequencing technologies facilitating the discovery of novel fusion events at an accelerated pace. Oncofusions exert their effects through the manipulation of critical cellular signaling pathways that regulate processes such as proliferation, differentiation, and survival. Extensive investigations have been conducted to understand the roles of oncofusions in solid tumors, leukemias, and lymphomas. Large-scale initiatives, including the Cancer Genome Atlas, have played a pivotal role in unraveling the landscape of oncofusions by characterizing a vast number of cancer samples across different tumor types. While validating the functional relevance of oncofusions remains a challenge, even non-driver mutations can hold significance in cancer treatment. Oncofusions have demonstrated potential value in the context of immunotherapy through the production of neoantigens. Their clinical importance has been observed in both treatment and diagnostic settings, with specific fusion events serving as therapeutic targets or diagnostic markers. However, despite the progress made, there is still considerable untapped potential within the field of oncofusions. Further research and validation efforts are necessary to understand their effects on a functional basis and to exploit the new targeted treatment avenues offered by oncofusions. Through further functional and clinical studies, oncofusions will enable the advancement of precision medicine and the drive towards more effective and specific treatments for cancer patients.
Collapse
Affiliation(s)
- Kari Salokas
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00790 Helsinki, Finland
| | - Giovanna Dashi
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00790 Helsinki, Finland
| | - Markku Varjosalo
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00790 Helsinki, Finland
| |
Collapse
|
27
|
Nitulescu GM, Stancov G, Seremet OC, Nitulescu G, Mihai DP, Duta-Bratu CG, Barbuceanu SF, Olaru OT. The Importance of the Pyrazole Scaffold in the Design of Protein Kinases Inhibitors as Targeted Anticancer Therapies. Molecules 2023; 28:5359. [PMID: 37513232 PMCID: PMC10385367 DOI: 10.3390/molecules28145359] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
The altered activation or overexpression of protein kinases (PKs) is a major subject of research in oncology and their inhibition using small molecules, protein kinases inhibitors (PKI) is the best available option for the cure of cancer. The pyrazole ring is extensively employed in the field of medicinal chemistry and drug development strategies, playing a vital role as a fundamental framework in the structure of various PKIs. This scaffold holds major importance and is considered a privileged structure based on its synthetic accessibility, drug-like properties, and its versatile bioisosteric replacement function. It has proven to play a key role in many PKI, such as the inhibitors of Akt, Aurora kinases, MAPK, B-raf, JAK, Bcr-Abl, c-Met, PDGFR, FGFRT, and RET. Of the 74 small molecule PKI approved by the US FDA, 8 contain a pyrazole ring: Avapritinib, Asciminib, Crizotinib, Encorafenib, Erdafitinib, Pralsetinib, Pirtobrutinib, and Ruxolitinib. The focus of this review is on the importance of the unfused pyrazole ring within the clinically tested PKI and on the additional required elements of their chemical structures. Related important pyrazole fused scaffolds like indazole, pyrrolo[1,2-b]pyrazole, pyrazolo[4,3-b]pyridine, pyrazolo[1,5-a]pyrimidine, or pyrazolo[3,4-d]pyrimidine are beyond the subject of this work.
Collapse
Affiliation(s)
| | | | | | - Georgiana Nitulescu
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania; (G.M.N.)
| | | | | | | | | |
Collapse
|
28
|
Xu Y, Wang Z, Zhang L, Gao C, Li F, Li X, Ke Y, Liu HM, Hu Z, Wei L, Chen ZS. Differentiation of imatinib -resistant chronic myeloid leukemia cells with BCR-ABL-T315I mutation induced by Jiyuan Oridonin A. J Cancer 2023; 14:1182-1194. [PMID: 37215441 PMCID: PMC10197941 DOI: 10.7150/jca.83219] [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] [Received: 02/04/2023] [Accepted: 04/19/2023] [Indexed: 05/24/2023] Open
Abstract
Chronic myeloid leukemia (CML) results from BCR-ABL oncogene, which blocks CML cells differentiation and protects these cells from apoptosis. T315I mutated BCR-ABL is the main cause of the resistance mediated by imatinib and second generation BCR-ABL inhibitor. CML with the T315I mutation has been considered to have poor prognosis. Here, we determined the effect of Jiyuan oridonin A (JOA), an ent-kaurene diterpenoid compound, on the differentiation blockade in imatinib-sensitive, particularly, imatinib-resistant CML cells with BCR-ABL-T315I mutation by cell proliferation assay, apoptosis analysis, cell differentiation analysis, cell cycle analysis and colony formation assay. We also investigated the possible molecular mechanism by mRNA sequencing, qRT-PCR and Western blotting. We found that JOA at lower concentration significantly inhibited the proliferation of CML cells expressing mutant BCR-ABL (T315I mutation included) and wild-type BCR-ABL, which was due to that JOA induced the cell differentiation and the cell cycle arrest at G0/G1 phase. Interestingly, JOA possessed stronger anti-leukemia activity than its analogues such as OGP46 and Oridonin, which has been investigated extensively. Mechanistically, the cell differentiation mediated by JOA may be originated from the inhibition of BCR-ABL/c-MYC signaling in CML cells expressing wild-type BCR-ABL and BCR-ABL-T315I. JOA displayed the activity of inhibiting the BCR-ABL and promoted differentiation of not only imatinib -sensitive but also imatinib -resistant cells with BCR-ABL mutation, which could become a potent lead compound to overcome the imatinib -resistant induced by inhibitors of BCR-ABL tyrosine kinase in CML therapy.
Collapse
Affiliation(s)
- Yun Xu
- School of Pharmacy, Weifang Medical University, Weifang, 261053, China
| | - Ziting Wang
- School of Pharmacy, Weifang Medical University, Weifang, 261053, China
| | - Lei Zhang
- School of Pharmacy, Weifang Medical University, Weifang, 261053, China
| | - Congying Gao
- School of Pharmacy, Weifang Medical University, Weifang, 261053, China
| | - Fahui Li
- School of Pharmacy, Weifang Medical University, Weifang, 261053, China
| | - Xueming Li
- School of Pharmacy, Weifang Medical University, Weifang, 261053, China
| | - Yu Ke
- School of Pharmacy, Zhengzhou University, Zhengzhou, 450052, China
| | - Hong-Min Liu
- School of Pharmacy, Zhengzhou University, Zhengzhou, 450052, China
| | - Zhenbo Hu
- Laboratory for Stem Cell and Regenerative Medicine, Affiliated Hospital of Weifang Medical University, Weifang 261042, China
| | - Liuya Wei
- School of Pharmacy, Weifang Medical University, Weifang, 261053, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| |
Collapse
|
29
|
Cereblon-Recruiting PROTACs: Will New Drugs Have to Face Old Challenges? Pharmaceutics 2023; 15:pharmaceutics15030812. [PMID: 36986673 PMCID: PMC10053963 DOI: 10.3390/pharmaceutics15030812] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
The classical low-molecular-weight drugs are designed to bind with high affinity to the biological targets endowed with receptor or enzymatic activity, and inhibit their function. However, there are many non-receptor or non-enzymatic disease proteins that seem undruggable using the traditional drug approach. This limitation has been overcome by PROTACs, bifunctional molecules that are able to bind the protein of interest and the E3 ubiquitin ligase complex. This interaction results in the ubiquitination of POI and subsequent proteolysis in the cellular proteasome. Out of hundreds of proteins serving as substrate receptors in E3 ubiquitin ligase complexes, current PROTACs recruit only a few of them, including CRBN, cIAP1, VHL or MDM-2. This review will focus on PROTACs recruiting CRBN E3 ubiquitin ligase and targeting various proteins involved in tumorigenesis, such as transcription factors, kinases, cytokines, enzymes, anti-apoptotic proteins and cellular receptors. The structure of several PROTACs, their chemical and pharmacokinetic properties, target affinity and biological activity in vitro and in vivo, will be discussed. We will also highlight cellular mechanisms that may affect the efficacy of PROTACs and pose a challenge for the future development of PROTACs.
Collapse
|
30
|
Kamyabi R, Jahandideh A, Panahi N, Muhammadnejad S. Synergistic cytotoxicity effect of the combination of chitosan nanoencapsulated imatinib mesylate and quercetin in BCR-ABL positive K562 cells. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:359-366. [PMID: 36865043 PMCID: PMC9922367 DOI: 10.22038/ijbms.2023.68472.14934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 12/18/2022] [Indexed: 03/04/2023]
Abstract
Objectives Intolerable side effects and resistance to chemotherapeutic drugs have encouraged scientists to develop new methods of drug combinations with fewer complications. This study aimed to investigate the synergistic effects of quercetin and imatinib encapsulated in chitosan nanoparticles on cytotoxicity, apoptosis, and cell growth of the K562 cell line. Materials and Methods Imatinib and quercetin were encapsulated in chitosan nanoparticles and their physical properties were determined using standard methods and SEM microscope images. BCR-ABL positive K562 cells were cultured in a cell culture medium, cytotoxicity of drugs was determined by MTT assay and the effects of nano drugs on apoptosis in cells were investigated by Annexin V-FITC staining. The expression level of genes associated with apoptosis in cells was measured by real-time PCR. Results The IC50 for the combination of the nano drugs at 24 and 48 hr was 9.324 and 10.86 μg/ml, respectively. The data indicated that the encapsulated form of drugs induced apoptosis more effectively than the free form (P<0.05). Moreover, the synergistic effect of nano drugs in statistical analysis was proved (P=0.001). The combination of nano drugs resulted in the caspase 3, 8, and TP53 genes upregulation (P=0.001). Conclusion The results of the present study showed that the encapsulated form of imatinib and quercetin nano drugs with chitosan has more cytotoxicity than the free form of the drugs. In addition, the combination of imatinib and quercetin as a nano-drug complex has a synergistic effect on the induction of apoptosis in imatinib-resistant K562 cells.
Collapse
Affiliation(s)
- Rohollah Kamyabi
- Department of Veterinary Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Alireza Jahandideh
- Department of Veterinary Surgery, Science and Research Branch, Islamic Azad University, Tehran, Iran,Corresponding author: Alireza Jahandideh. Department of Veterinary Surgery, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Negar Panahi
- Department of Veterinary Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Samad Muhammadnejad
- Gene Therapy Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Cao X, Jin X, Zhang X, Utsav P, Zhang Y, Guo R, Lu W, Zhao M. Small-Molecule Compounds Boost CAR-T Cell Therapy in Hematological Malignancies. Curr Treat Options Oncol 2023; 24:184-211. [PMID: 36701037 PMCID: PMC9992085 DOI: 10.1007/s11864-023-01049-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2022] [Indexed: 01/27/2023]
Abstract
OPINION STATEMENT Although chimeric antigen receptor T cell immunotherapy has been successfully applied in patients with hematological malignancies, several obstacles still need to be overcome, such as high relapse rates and side effects. Overcoming the limitations of CAR-T cell therapy and boosting the efficacy of CAR-T cell therapy are urgent issues that must be addressed. The exploration of small-molecule compounds in combination with CAR-T cell therapies has achieved promising success in pre-clinical and clinical studies in recent years. Protein kinase inhibitors, demethylating drugs, HDAC inhibitors, PI3K inhibitors, immunomodulatory drugs, Akt inhibitors, mTOR inhibitors, and Bcl-2 inhibitors exhibited potential synergy in combination with CAR-T cell therapy. In this review, we will discuss the recent application of these combination therapies for improved outcomes of CAR-T cell therapy.
Collapse
Affiliation(s)
- Xinping Cao
- First Center Clinic College of Tianjin Medical University, Tianjin, 300192, China
| | - Xin Jin
- Department of Hematology, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Xiaomei Zhang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Paudel Utsav
- First Center Clinic College of Tianjin Medical University, Tianjin, 300192, China
| | - Yi Zhang
- First Center Clinic College of Tianjin Medical University, Tianjin, 300192, China
| | - Ruiting Guo
- First Center Clinic College of Tianjin Medical University, Tianjin, 300192, China
| | - Wenyi Lu
- Department of Hematology, Tianjin First Central Hospital, Tianjin, 300192, China.
| | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, Tianjin, 300192, China.
| |
Collapse
|
32
|
BH3 mimetics and TKI combined therapy for Chronic Myeloid Leukemia. Biochem J 2023; 480:161-176. [PMID: 36719792 DOI: 10.1042/bcj20210608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 02/01/2023]
Abstract
Chronic myeloid leukemia (CML) was considered for a long time one of the most hostile leukemia that was incurable for most of the patients, predominantly due to the extreme resistance to chemotherapy. Part of the resistance to cell death (apoptosis) is the result of increased levels of anti-apoptotic and decreased levels of pro-apoptotic member of the BCL-2 family induced by the BCR-ABL1 oncoprotein. BCR-ABL1 is a constitutively active tyrosine kinase responsible for initiating multiple and oncogenic signaling pathways. With the development of specific BCR-ABL1 tyrosine kinase inhibitors (TKIs) CML became a much more tractable disease. Nevertheless, TKIs do not cure CML patients and a substantial number of them develop intolerance or become resistant to the treatment. Therefore, novel anti-cancer strategies must be developed to treat CML patients independently or in combination with TKIs. Here, we will discuss the mechanisms of BCR-ABL1-dependent and -independent resistance to TKIs and the use of BH3-mimetics as a potential tool to fight CML.
Collapse
|
33
|
Vicente ATS, Salvador JAR. Proteolysis-Targeting Chimeras (PROTACs) targeting the BCR-ABL for the treatment of chronic myeloid leukemia - a patent review. Expert Opin Ther Pat 2023; 33:397-420. [PMID: 37494069 DOI: 10.1080/13543776.2023.2240025] [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] [Received: 03/07/2023] [Accepted: 07/19/2023] [Indexed: 07/27/2023]
Abstract
INTRODUCTION PROteolysis-TArgeting Chimeras (PROTACs) allow the selective degradation of a protein of interest (POI) by the ubiquitin-proteasome system (UPS). With this unique mechanism of action, the research and development of PROTACs that target the Breakpoint Cluster Region Abelson (BCR-ABL) tyrosine kinase (TK) has been increasing dramatically, as they are promising molecules in the treatment of Chronic Myeloid Leukemia (CML), one of the main hematological malignancies, which results from an uncontrolled myeloproliferation due to the constitutive activation of BCR-ABL. AREAS COVERED This review summarizes the patents/applications published in the online databases like Espacenet or World Intellectual Property Organization regarding PROTACs that promote BCR-ABL degradation. Patents will be described mostly in terms of chemical structure, biochemical/pharmacological activities, and potential clinical applications. EXPERT OPINION The recent discovery of the enormous potential of PROTACs led to the creation of new compounds capable of degrading BCR-ABL for the treatment of CML. Although still in reduced numbers, and in the pre-clinical phase of development, some compounds have already been shown to overcome some of the difficulties presented by conventional BCR-ABL inhibitors, such as the well-known imatinib. Therefore, it is very likely that some of the present PROTACs will enter future CML therapy in the coming years.
Collapse
MESH Headings
- Humans
- Proteolysis Targeting Chimera
- Proteolysis
- Drug Resistance, Neoplasm
- Patents as Topic
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Fusion Proteins, bcr-abl/chemistry
- Fusion Proteins, bcr-abl/metabolism
- Protein Kinase Inhibitors/chemistry
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
Collapse
Affiliation(s)
- André T S Vicente
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Jorge A R Salvador
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology (CNC), Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
34
|
Sorokin M, Rabushko E, Rozenberg JM, Mohammad T, Seryakov A, Sekacheva M, Buzdin A. Clinically relevant fusion oncogenes: detection and practical implications. Ther Adv Med Oncol 2022; 14:17588359221144108. [PMID: 36601633 PMCID: PMC9806411 DOI: 10.1177/17588359221144108] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 11/22/2022] [Indexed: 12/28/2022] Open
Abstract
Mechanistically, chimeric genes result from DNA rearrangements and include parts of preexisting normal genes combined at the genomic junction site. Some rearranged genes encode pathological proteins with altered molecular functions. Those which can aberrantly promote carcinogenesis are called fusion oncogenes. Their formation is not a rare event in human cancers, and many of them were documented in numerous study reports and in specific databases. They may have various molecular peculiarities like increased stability of an oncogenic part, self-activation of tyrosine kinase receptor moiety, and altered transcriptional regulation activities. Currently, tens of low molecular mass inhibitors are approved in cancers as the drugs targeting receptor tyrosine kinase (RTK) oncogenic fusion proteins, that is, including ALK, ABL, EGFR, FGFR1-3, NTRK1-3, MET, RET, ROS1 moieties. Therein, the presence of the respective RTK fusion in the cancer genome is the diagnostic biomarker for drug prescription. However, identification of such fusion oncogenes is challenging as the breakpoint may arise in multiple sites within the gene, and the exact fusion partner is generally unknown. There is no gold standard method for RTK fusion detection, and many alternative experimental techniques are employed nowadays to solve this issue. Among them, RNA-seq-based methods offer an advantage of unbiased high-throughput analysis of only transcribed RTK fusion genes, and of simultaneous finding both fusion partners in a single RNA-seq read. Here we focus on current knowledge of biology and clinical aspects of RTK fusion genes, related databases, and laboratory detection methods.
Collapse
Affiliation(s)
| | - Elizaveta Rabushko
- Moscow Institute of Physics and Technology,
Dolgoprudny, Moscow Region, Russia,I.M. Sechenov First Moscow State Medical
University, Moscow, Russia
| | | | - Tharaa Mohammad
- Moscow Institute of Physics and Technology,
Dolgoprudny, Moscow Region, Russia
| | | | - Marina Sekacheva
- I.M. Sechenov First Moscow State Medical
University, Moscow, Russia
| | - Anton Buzdin
- Moscow Institute of Physics and Technology,
Dolgoprudny, Moscow Region, Russia,I.M. Sechenov First Moscow State Medical
University, Moscow, Russia,Shemyakin-Ovchinnikov Institute of Bioorganic
Chemistry, Moscow, Russia,PathoBiology Group, European Organization for
Research and Treatment of Cancer (EORTC), Brussels, Belgium
| |
Collapse
|
35
|
Liu H, Mi Q, Ding X, Lin C, Liu L, Ren C, Shen S, Shao Y, Chen J, Zhou Y, Ji L, Zhang H, Bai F, Yang X, Yin Q, Jiang B. Discovery and characterization of novel potent BCR-ABL degraders by conjugating allosteric inhibitor. Eur J Med Chem 2022; 244:114810. [DOI: 10.1016/j.ejmech.2022.114810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/04/2022]
|
36
|
Elderdery AY, Alzahrani B, Hamza SMA, Mostafa-Hedeab G, Mok PL, Subbiah SK. CuO-TiO 2-Chitosan-Berbamine Nanocomposites Induce Apoptosis through the Mitochondrial Pathway with the Expression of P53, BAX, and BCL-2 in the Human K562 Cancer Cell Line. Bioinorg Chem Appl 2022; 2022:9602725. [PMID: 36164585 PMCID: PMC9509271 DOI: 10.1155/2022/9602725] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/18/2022] [Accepted: 07/06/2022] [Indexed: 11/17/2022] Open
Abstract
In this study, cells from human Chronic Myelogenous Leukemia (K562) were cultivated with CuO-TiO2-Chitosan-Berbamine nanocomposites. We examined nanocomposites using XRD, DLS, FESEM, TEM, PL, EDAX, and FTIR spectroscopy, as well as MTT for cytotoxicity, and AO/EtBr for apoptotic morphology assessment. The rate of apoptosis and cell cycle arrests was determined using flow cytometry. Flow cytometry was also employed to identify pro- and antiapoptotic proteins such as Bcl2, Bad, Bax, P53, and Cyt C. The FTIR spectrum revealed that the CuO-TiO2-Chitosan-Berbamine nanocomposites were electrostatically interlocked. The nanocomposites' XRD signals revealed a hexagonal shape. In the DLS spectrum, nanocomposites were found to have a hydrodynamic diameter. As a result of their cytotoxic action, nanocomposites displayed concentration-dependent cytotoxicity. The nanocomposites, like Doxorubicin, caused cell cycle phase arrest in K562 cells. After treatment with IC50 concentrations of CuO-TiO2-Chitosan-Berbamine nanocomposites and Doxorubicin, a substantial percentage of cells were in G2/M stage arrest. Caspase-3, -7, -8, -9, Bax, Bad, Cyt C, and P53 expression were considerably enhanced in K562 cells, whereas Bcl2 expression was decreased, indicating that these cells may have therapeutic potential against human blood cancer/leukemia-derived disorders. As a result, the nanocomposites demonstrated outstanding anticancer potential against leukemic cells. CuO-TiO2-Chitosan-Berbamine, according to our findings.
Collapse
Affiliation(s)
- Abozer Y. Elderdery
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Badr Alzahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Siddiqa M. A Hamza
- Faculty of Medicine, Department of Pathology, Umm Alqura University, Algunfuda, Mecca, Saudi Arabia
| | - Gomaa Mostafa-Hedeab
- Pharmacology & Therapeutic Department-Medical College, Jouf University, Sakaka, Saudi Arabia
| | - Pooi Ling Mok
- Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Suresh Kumar Subbiah
- Centre for Materials Engineering and Regenerative Medicine, Bharath Institute of Higher Education and Research, Chennai, India
| |
Collapse
|
37
|
Daniel JP, Mesquita FP, Da Silva EL, de Souza PFN, Lima LB, de Oliveira LLB, de Moraes MEA, Moreira-Nunes CDFA, Burbano RMR, Zanatta G, Montenegro RC. Anticancer potential of mebendazole against chronic myeloid leukemia: in silico and in vitro studies revealed new insights about the mechanism of action. Front Pharmacol 2022; 13:952250. [PMID: 36091760 PMCID: PMC9452629 DOI: 10.3389/fphar.2022.952250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic myeloid leukemia (CML) is caused by constitutively active fusion protein BCR-ABL1, and targeting ABL1 is a promising therapy option. Imatinib, dasatinib, and nilotinib have all been shown to work effectively in clinical trials. ABL1 mutations, particularly the T315I gate-keeper mutation, cause resistance in patients. As a result, broad-spectrum ABL1 medicines are desperately needed. In order to screen potential drugs targeting CML, mebendazole (MBZ) was subjected to the in vitro test against CML cell lines (K562 and FEPS) and computational assays. The antiproliferative effect of MBZ and the combination with tyrosine kinase inhibitors (TKIs) was tested using end-point viability assays, cell cycle distribution analysis, cell membrane, and mitochondrial dyes. By interrupting the cell cycle and causing cell death, MBZ and its combination with imatinib and dasatinib have a significant antiproliferative effect. We identified MBZ as a promising “new use” drug targeting wild-type and mutant ABL1 using molecular docking. Meanwhile, we determined which residues in the allosteric site are important in ABL1 drug development. These findings may not only serve as a model for repositioning current authorized medications but may also provide ABL1-targeted anti-CML treatments a fresh lease of life.
Collapse
Affiliation(s)
- Julio Paulino Daniel
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | - Felipe Pantoja Mesquita
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | - Emerson Lucena Da Silva
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | - Pedro Filho Noronha de Souza
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
- Department of Biochemistry and Molecular Biology, Federal University of Ceará, Fortaleza, Brazil
| | - Luina Benevides Lima
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | | | | | - Caroline de Fátima Aquino Moreira-Nunes
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém, Brazil
| | - Rommel Mario Rodríguez Burbano
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém, Brazil
- Molecular Biology Laboratory, Ophir Loyola Hospital, Belém, Brazil
| | - Geancarlo Zanatta
- Department of Physics, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Raquel Carvalho Montenegro
- Laboratory of Pharmacogenetics, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
- *Correspondence: Raquel Carvalho Montenegro,
| |
Collapse
|
38
|
Stukan I, Gryzik M, Hoser G, Want A, Grabowska-Pyrzewicz W, Zdioruk M, Napiórkowska M, Cieślak M, Królewska-Golińska K, Nawrot B, Basak G, Wojda U. Novel Dicarboximide BK124.1 Breaks Multidrug Resistance and Shows Anticancer Efficacy in Chronic Myeloid Leukemia Preclinical Models and Patients' CD34 +/CD38 - Leukemia Stem Cells. Cancers (Basel) 2022; 14:cancers14153641. [PMID: 35892900 PMCID: PMC9332833 DOI: 10.3390/cancers14153641] [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] [Received: 06/10/2022] [Revised: 07/20/2022] [Accepted: 07/23/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Chemotherapy is a first line treatment in many cancer types, but the constant exposition to chemotherapeutics often leads to therapy resistance. An example is chronic myeloid leukemia that, due to the use of tyrosine kinase inhibitors such as imatinib, remains manageable, however incurable. Overall, 20–25% of imatinib responders develop secondary resistance, and among them, 20–40% is due to mechanisms such as expression of P-glycoprotein (MDR1) or leukemia stem cells’ mechanisms of survival and cancer regrowth. This study provides the first evidence from animal and cellular models that this resistance can be overcome with the novel dicarboximide BK124.1. The compound causes no visible toxicity in mice, and has proper pharmacokinetics for therapeutic applications. It was efficient against both multidrug resistant CML blasts and CD34+/CD38− leukemia stem cells coming from CML patients. Future development of BK124.1 could offer curative treatment of CML and of other cancers resistant or intolerant to current chemotherapy. Abstract The search is ongoing for new anticancer therapeutics that would overcome resistance to chemotherapy. This includes chronic myeloid leukemia, particularly suitable for the studies of novel anticancer compounds due to its homogenous and well-known genetic background. Here we show anticancer efficacy of novel dicarboximide denoted BK124.1 (C31H37ClN2O4) in a mouse CML xenograft model and in vitro in two types of chemoresistant CML cells: MDR1 blasts and in CD34+ patients’ stem cells (N = 8) using immunoblotting and flow cytometry. Intraperitoneal administration of BK124.1 showed anti-CML efficacy in the xenograft mouse model (N = 6) comparable to the commonly used imatinib and hydroxyurea. In K562 blasts, BK124.1 decreased the protein levels of BCR-ABL1 kinase and its downstream effectors, resulting in G2/M cell cycle arrest and apoptosis associated with FOXO3a/p21waf1/cip1 upregulation in the nucleus. Additionally, BK124.1 evoked massive apoptosis in multidrug resistant K562-MDR1 cells (IC50 = 2.16 μM), in CD34+ cells from CML patients (IC50 = 1.5 µM), and in the CD34+/CD38− subpopulation consisting of rare, drug-resistant cancer initiating stem cells. Given the advantages of BK124.1 as a potential chemotherapeutic and its unique ability to overcome BCR-ABL1 dependent and independent multidrug resistance mechanisms, future development of BK124.1 could offer a cure for CML and other cancers resistant to present drugs.
Collapse
Affiliation(s)
- Iga Stukan
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (I.S.); (M.G.); (G.H.); (A.W.); (W.G.-P.); (M.Z.)
| | - Marek Gryzik
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (I.S.); (M.G.); (G.H.); (A.W.); (W.G.-P.); (M.Z.)
| | - Grażyna Hoser
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (I.S.); (M.G.); (G.H.); (A.W.); (W.G.-P.); (M.Z.)
- Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
| | - Andrew Want
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (I.S.); (M.G.); (G.H.); (A.W.); (W.G.-P.); (M.Z.)
| | - Wioleta Grabowska-Pyrzewicz
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (I.S.); (M.G.); (G.H.); (A.W.); (W.G.-P.); (M.Z.)
| | - Mikolaj Zdioruk
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (I.S.); (M.G.); (G.H.); (A.W.); (W.G.-P.); (M.Z.)
| | - Mariola Napiórkowska
- Department of Biochemistry, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Marcin Cieślak
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 90-363 Lodz, Poland; (M.C.); (K.K.-G.); (B.N.)
| | - Karolina Królewska-Golińska
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 90-363 Lodz, Poland; (M.C.); (K.K.-G.); (B.N.)
| | - Barbara Nawrot
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, 90-363 Lodz, Poland; (M.C.); (K.K.-G.); (B.N.)
| | - Grzegorz Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Urszula Wojda
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (I.S.); (M.G.); (G.H.); (A.W.); (W.G.-P.); (M.Z.)
- Correspondence: ; Tel.: +48-22-5892-578
| |
Collapse
|
39
|
Poudel G, Tolland MG, Hughes TP, Pagani IS. Mechanisms of Resistance and Implications for Treatment Strategies in Chronic Myeloid Leukaemia. Cancers (Basel) 2022; 14:cancers14143300. [PMID: 35884363 PMCID: PMC9317051 DOI: 10.3390/cancers14143300] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/28/2022] [Accepted: 07/04/2022] [Indexed: 12/01/2022] Open
Abstract
Simple Summary Chronic myeloid leukaemia (CML) is a type of blood cancer that is currently well-managed with drugs that target cancer-causing proteins. However, a significant proportion of CML patients do not respond to those drug treatments or relapse when they stop those drugs because the cancer cells in those patients stop relying on that protein and instead develop a new way to survive. Therefore, new treatment strategies may be necessary for those patients. In this review, we discuss those additional survival pathways and outline combination treatment strategies to increase responses and clinical outcomes, improving the lives of CML patients. Abstract Tyrosine kinase inhibitors (TKIs) have revolutionised the management of chronic myeloid leukaemia (CML), with the disease now having a five-year survival rate over 80%. The primary focus in the treatment of CML has been on improving the specificity and potency of TKIs to inhibit the activation of the BCR::ABL1 kinase and/or overcoming resistance driven by mutations in the BCR::ABL1 oncogene. However, this approach may be limited in a significant proportion of patients who develop TKI resistance despite the effective inhibition of BCR::ABL1. These patients may require novel therapeutic strategies that target both BCR::ABL1-dependent and BCR::ABL1-independent mechanisms of resistance. The combination treatment strategies that target alternative survival signalling, which may contribute towards BCR::ABL1-independent resistance, could be a successful strategy for eradicating residual leukaemic cells and consequently increasing the response rate in CML patients.
Collapse
Affiliation(s)
- Govinda Poudel
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia; (G.P.); (M.G.T.); (T.P.H.)
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Australasian Leukaemia and Lymphoma Group, Richmond, VIC 3121, Australia
| | - Molly G. Tolland
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia; (G.P.); (M.G.T.); (T.P.H.)
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
| | - Timothy P. Hughes
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia; (G.P.); (M.G.T.); (T.P.H.)
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Australasian Leukaemia and Lymphoma Group, Richmond, VIC 3121, Australia
- Department of Haematology and Bone Marrow Transplantation, Royal Adelaide Hospital and SA Pathology, Adelaide, SA 5000, Australia
| | - Ilaria S. Pagani
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia; (G.P.); (M.G.T.); (T.P.H.)
- School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia
- Australasian Leukaemia and Lymphoma Group, Richmond, VIC 3121, Australia
- Correspondence:
| |
Collapse
|
40
|
Targeting SAMHD1: to overcome multiple anti-cancer drugs resistance in hematological malignancies. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
41
|
Chromosomal Instability in Chronic Myeloid Leukemia: Mechanistic Insights and Effects. Cancers (Basel) 2022; 14:cancers14102533. [PMID: 35626137 PMCID: PMC9140097 DOI: 10.3390/cancers14102533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 12/15/2022] Open
Abstract
The most recent two decades have seen tremendous progress in the understanding and treatment of chronic myeloid leukemia, a disease defined by the characteristic Philadelphia chromosome and the ensuing BCR::ABL fusion protein. However, the biology of the disease extends beyond the Philadelphia chromosome into a nebulous arena of chromosomal and genetic instability, which makes it a genetically heterogeneous disease. The BCR::ABL oncoprotein creates a fertile backdrop for oxidative damage to the DNA, along with impairment of genetic surveillance and the favoring of imprecise error-prone DNA repair pathways. These factors lead to growing chromosomal instability, manifested as additional chromosomal abnormalities along with other genetic aberrations. This worsens with disease progression to accelerated and blast phase, and modulates responses to tyrosine kinase inhibitors. Treatment options that target the genetic aberrations that mitigate chromosome instability might be a potential area for research in patients with advanced phase CML.
Collapse
|
42
|
Dactylospongia elegans—A Promising Drug Source: Metabolites, Bioactivities, Biosynthesis, Synthesis, and Structural-Activity Relationship. Mar Drugs 2022; 20:md20040221. [PMID: 35447894 PMCID: PMC9033123 DOI: 10.3390/md20040221] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/17/2022] [Accepted: 03/22/2022] [Indexed: 02/07/2023] Open
Abstract
Marine environment has been identified as a huge reservoir of novel biometabolites that are beneficial for medical treatments, as well as improving human health and well-being. Sponges have been highlighted as one of the most interesting phyla as new metabolites producers. Dactylospongia elegans Thiele (Thorectidae) is a wealth pool of various classes of sesquiterpenes, including hydroquinones, quinones, and tetronic acid derivatives. These metabolites possessed a wide array of potent bioactivities such as antitumor, cytotoxicity, antibacterial, and anti-inflammatory. In the current work, the reported metabolites from D. elegans have been reviewed, including their bioactivities, biosynthesis, and synthesis, as well as the structural-activity relationship studies. Reviewing the reported studies revealed that these metabolites could contribute to new drug discovery, however, further mechanistic and in vivo studies of these metabolites are needed.
Collapse
|
43
|
Sasaki K, Fujiwara T, Ochi T, Ono K, Kato H, Onodera K, Ichikawa S, Fukuhara N, Onishi Y, Yokoyama H, Miyata T, Harigae H. TM5614, an Inhibitor of Plasminogen Activator Inhibitor-1, Exerts an Antitumor Effect on Chronic Myeloid Leukemia. TOHOKU J EXP MED 2022; 257:211-224. [DOI: 10.1620/tjem.2022.j036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
| | - Tohru Fujiwara
- Department of Hematology, Tohoku University Graduate School
| | - Tetsuro Ochi
- Department of Hematology, Tohoku University Graduate School
| | - Koya Ono
- Department of Hematology, Tohoku University Graduate School
| | - Hiroki Kato
- Department of Hematology, Tohoku University Graduate School
| | - Koichi Onodera
- Department of Hematology, Tohoku University Graduate School
| | | | | | - Yasushi Onishi
- Department of Hematology, Tohoku University Graduate School
| | | | - Toshio Miyata
- Department of Molecular Medicine and Therapy, United Centers for Advanced Research and Translational Medicine
| | | |
Collapse
|