1
|
Pratiwi L, Mashudi FH, Ningtyas MC, Sutanto H, Romadhon PZ. Genetic Profiling of Acute and Chronic Leukemia via Next-Generation Sequencing: Current Insights and Future Perspectives. Hematol Rep 2025; 17:18. [PMID: 40277842 DOI: 10.3390/hematolrep17020018] [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: 03/04/2025] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025] Open
Abstract
Leukemia is a heterogeneous group of hematologic malignancies characterized by distinct genetic and molecular abnormalities. Advancements in genomic technologies have significantly transformed the diagnosis, prognosis, and treatment strategies for leukemia. Among these, next-generation sequencing (NGS) has emerged as a powerful tool, enabling high-resolution genomic profiling that surpasses conventional diagnostic approaches. By providing comprehensive insights into genetic mutations, clonal evolution, and resistance mechanisms, NGS has revolutionized precision medicine in leukemia management. Despite its transformative potential, the clinical integration of NGS presents challenges, including data interpretation complexities, standardization issues, and cost considerations. However, continuous advancements in sequencing platforms and bioinformatics pipelines are enhancing the reliability and accessibility of NGS in routine clinical practice. The expanding role of NGS in leukemia is paving the way for improved risk stratification, targeted therapies, and real-time disease monitoring, ultimately leading to better patient outcomes. This review highlights the impact of NGS on leukemia research and clinical applications, discussing its advantages over traditional diagnostic techniques, key sequencing approaches, and emerging challenges. As precision oncology continues to evolve, NGS is expected to play an increasingly central role in the diagnosis and management of leukemia, driving innovations in personalized medicine and therapeutic interventions.
Collapse
Affiliation(s)
- Laras Pratiwi
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Fawzia Hanum Mashudi
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Mukti Citra Ningtyas
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Henry Sutanto
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia
| | - Pradana Zaky Romadhon
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
- Department of Internal Medicine, Airlangga University Hospital, Surabaya 60115, Indonesia
| |
Collapse
|
2
|
Appasamy P, Nag JK, Malka H, Bar-Shavit R. PAR 2 Serves an Indispensable Role in Controlling PAR 4 Oncogenicity: The β-Catenin-p53 Axis. Int J Mol Sci 2025; 26:2780. [PMID: 40141421 PMCID: PMC11942634 DOI: 10.3390/ijms26062780] [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: 02/12/2025] [Revised: 03/18/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
Although the role of G-protein-coupled receptors (GPCRs) in cancer is acknowledged, GPCR-based cancer therapy is rare. Mammalian protease-activated receptors (PARs), a sub-group of GPCRs, comprise four family members, termed PAR1-4. Here, we demonstrate that PAR2 is dominant over PAR4 oncogene in cancer. We performed a knockdown of Par2/f2rl1 and expressed C-terminally truncated PAR2 (TrPAR2), incapable of inducing signaling, to assess the impact of PAR2 on PAR4 oncogenic function by β-catenin stabilization assessment, immunoprecipitation, and xenograft tumor generation in Nude/Nude mice. PAR2 and PAR4 act together to promote tumor generation. Knockdown Par2 and TrPAR2 inhibited the PAR2 and PAR4-induced β-catenin levels, nuclear dishevelled 1(DVL1), and TOPflash reporter activity. Likewise, PAR2 and PAR4-induced invasion and migration were inhibited when Par2 was knocked down or in the presence of TrPAR2. PAR cyclic (4-4) [Pc(4-4)], a PAR-based compound directed toward the PAR pleckstrin homology (PH)-binding site, effectively inhibited PAR2 oncogenic activity. Pc(4-4) inhibition is mediated via the increase in p53 level and the up-regulation of p21 as caspase-3 as well. Overall, we showed that in the absence of PAR2 signaling, the PAR4 pro-tumor functions are significantly inhibited. Pc(4-4) inhibits PAR2 acting via the modification of wt p53, thus offering a powerful drug measure for fighting cancer.
Collapse
Affiliation(s)
| | | | | | - Rachel Bar-Shavit
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (P.A.)
| |
Collapse
|
3
|
Wen W, Zhang WL, Tan R, Zhong TT, Zhang MR, Fang XS. Progress in deciphering the role of p53 in diffuse large B-cell lymphoma: mechanisms and therapeutic targets. Am J Cancer Res 2024; 14:3280-3293. [PMID: 39113862 PMCID: PMC11301306 DOI: 10.62347/lhio8294] [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: 01/31/2024] [Accepted: 05/30/2024] [Indexed: 08/10/2024] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoma subtype, accounting for 30%-40% of non-Hodgkin lymphoma in adults. The mechanisms underlying DLBCL occurrence are extremely complex, and involve the B-cell receptor (BCR) and Toll-like receptor (TLR) signaling pathways, as well as genetic abnormalities and other factors. With the development of high-throughput sequencing, an increasing number of abnormal genes have been identified in DLBCL. Among them, the tumor protein p53 (TP53/p53) gene is important in DLBCL occurrence. Patients with DLBCL carrying TP53 gene abnormalities generally have poor prognosis and studies of p53 have potential to provide a better basis for their treatment. Normally, p53 is maintained at low levels through its interaction with murine double minute 2 (MDM2), and prevents tumorigenesis by mediating cell cycle arrest, apoptosis, and repair of damaged cells, among other processes. Therefore, the prognosis of patients with DLBCL harboring TP53 gene abnormalities (mutations, deletions, etc.) is poor, and targeting p53 for tumor therapy has become a research hotspot, following developments in molecular biology technologies. Current treatments targeting p53 mainly act by restoring the function or promoting degradation of mutant p53, and enhancing wild-type p53 protein stability and activity. Based on the current status of p53 research, exploration of existing therapeutic methods to improve the prognosis of patients with DLBCL with TP53 abnormalities is warranted.
Collapse
Affiliation(s)
- Wen Wen
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinan 250021, Shandong, China
- Shandong First Medical UniversityJinan 250024, Shandong, China
| | - Wen-Lu Zhang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinan 250021, Shandong, China
- Shandong First Medical UniversityJinan 250024, Shandong, China
| | - Ran Tan
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinan 250021, Shandong, China
- Shandong First Medical UniversityJinan 250024, Shandong, China
| | - Tan-Tan Zhong
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinan 250021, Shandong, China
- Shandong First Medical UniversityJinan 250024, Shandong, China
| | - Mei-Rui Zhang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinan 250021, Shandong, China
- Shandong First Medical UniversityJinan 250024, Shandong, China
| | - Xiao-Sheng Fang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinan 250021, Shandong, China
- Shandong First Medical UniversityJinan 250024, Shandong, China
| |
Collapse
|
4
|
DiNardo CD, Olin R, Wang ES, Skikne B, Rosenthal J, Kumar P, Sumi H, Hizukuri Y, Hong Y, Patel P, Seki T, Duan T, Lesegretain A, Andreeff M. Phase 1 dose escalation study of the MDM2 inhibitor milademetan as monotherapy and in combination with azacitidine in patients with myeloid malignancies. Cancer Med 2024; 13:e70028. [PMID: 39030997 PMCID: PMC11258486 DOI: 10.1002/cam4.70028] [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: 02/29/2024] [Revised: 05/29/2024] [Accepted: 07/07/2024] [Indexed: 07/22/2024] Open
Abstract
BACKGROUND Mouse double minute-2 homolog (MDM2) plays a key role in downregulating p53 activity in hematologic malignancies, and its overexpression is associated with poor outcomes. METHODS This phase 1 study assessed the safety and efficacy of different dosing regimens of the MDM2 inhibitor milademetan as monotherapy and in combination with azacitidine (AZA) in patients with relapsed or refractory acute myeloid leukemia or high-risk myelodysplastic syndromes. RESULTS Seventy-four patients (monotherapy, n = 57; milademetan-AZA combination, n = 17) were treated. The maximum tolerated dose of milademetan was 160 mg once daily given for the first 14-21 days of 28-day cycles as monotherapy and on Days 5-14 in combination with AZA. Dose-limiting toxicities were gastrointestinal, fatigue, or renal/electrolyte abnormalities. Treatment-emergent adverse events related to milademetan occurred in 82.5% and 64.7% of participants in the monotherapy and AZA combination arms, respectively. Two participants (4.2%) in the monotherapy arm achieved complete remission (CR), and 1 (2.1%) achieved CR with incomplete blood count recovery (CRi). Two participants (13.3%) achieved CRi in the combination arm. New TP53 mutations, detected only during milademetan monotherapy, were found pre-existing below standard detection frequency by droplet digital polymerase chain reaction. INTERPRETATION Milademetan was relatively well tolerated in this population; however, despite signals of activity, clinical efficacy was minimal.
Collapse
Affiliation(s)
- Courtney D. DiNardo
- Department of LeukemiaThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Rebecca Olin
- University of CaliforniaSan FranciscoCaliforniaUSA
| | - Eunice S. Wang
- Roswell Park Comprehensive Care CenterBuffaloNew YorkUSA
| | - Barry Skikne
- University of Kansas Medical CenterKansas CityKansasUSA
| | | | | | | | | | - Ying Hong
- Daiichi Sankyo Inc.Basking RidgeNew JerseyUSA
| | - Parul Patel
- Daiichi Sankyo Inc.Basking RidgeNew JerseyUSA
| | | | - Tao Duan
- Daiichi Sankyo Inc.Basking RidgeNew JerseyUSA
| | | | - Michael Andreeff
- Department of LeukemiaThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| |
Collapse
|
5
|
Wang K, Jiang M, Liu H, Meng C, Li M, Lu H. Discovery of novel co-degradation CK1α and CDK7/9 PROTACs with p53 activation for treating acute myeloid leukemia. Bioorg Chem 2024; 147:107319. [PMID: 38593529 DOI: 10.1016/j.bioorg.2024.107319] [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: 01/14/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/11/2024]
Abstract
Reactivating p53 activity to restore its anticancer function is an attractive cancer treatment strategy. In this study, we designed and synthesized a series of novel PROTACs to reactivate p53 via the co-degradation of CK1α and CDK7/9 proteins. Bioactivity studies showed that the selected PROTAC 13i exhibited potency antiproliferative activity in MV4-11 (IC50 = 0.096 ± 0.012 μM) and MOLM-13 (IC50 = 0.072 ± 0.014 μM) cells, and induced apoptosis of MV4-11 cells. Western-blot analysis showed that PROTAC 13i triple CK1α and CDK7/9 protein degradation resulted in the significantly increased expression of p53. At the same time, the transcriptional repression due to the degradation significantly reduced downstream gene expression of MYC, MDM2, BCL-2 and MCL-1, and reduced the inflammatory cytokine levels of TNF-α, IL-1β and IL-6 in PMBCs. These results indicate the beneficial impact of simultaneous CK1α and CDK7/9 degradation for acute myeloid leukemia therapy.
Collapse
MESH Headings
- Humans
- Tumor Suppressor Protein p53/metabolism
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/chemical synthesis
- Casein Kinase Ialpha/metabolism
- Casein Kinase Ialpha/antagonists & inhibitors
- Cell Proliferation/drug effects
- Drug Screening Assays, Antitumor
- Cyclin-Dependent Kinase 9/antagonists & inhibitors
- Cyclin-Dependent Kinase 9/metabolism
- Structure-Activity Relationship
- Molecular Structure
- Cyclin-Dependent Kinases/antagonists & inhibitors
- Cyclin-Dependent Kinases/metabolism
- Dose-Response Relationship, Drug
- Apoptosis/drug effects
- Drug Discovery
- Cell Line, Tumor
- Proteolysis/drug effects
- Tumor Cells, Cultured
- Proteolysis Targeting Chimera
- Cyclin-Dependent Kinase-Activating Kinase
Collapse
Affiliation(s)
- Kai Wang
- College of Pharmacy, Jilin University, Changchun 130021, China
| | - Meixu Jiang
- College of Pharmacy, Jilin University, Changchun 130021, China
| | - Huimin Liu
- College of Pharmacy, Jilin University, Changchun 130021, China
| | - Chen Meng
- College of Pharmacy, Jilin University, Changchun 130021, China
| | - Mengyuan Li
- College of Pharmacy, Jilin University, Changchun 130021, China
| | - Haibin Lu
- College of Pharmacy, Jilin University, Changchun 130021, China.
| |
Collapse
|
6
|
Shomali N, Kamrani A, Nasiri H, Heris JA, Shahabi P, Yousefi M, Mohammadinasab R, Sadeghvand S, Akbari M. An updated review of a novel method for examining P53 mutations in different forms of cancer. Pathol Res Pract 2023; 248:154585. [PMID: 37302277 DOI: 10.1016/j.prp.2023.154585] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/27/2023] [Accepted: 05/28/2023] [Indexed: 06/13/2023]
Abstract
In the past fifteen years, it has been clear that tumor-associated p53 mutations can cause behaviors distinct from those brought on by a simple loss of p53's tumor-suppressive function in its wild-type form. Many of these mutant p53 proteins develop oncogenic characteristics that allow them to encourage cell survival, invasion, and metastasis. But it is now understood that the immune response is also significantly influenced by the cancer cell's p53 status. The recruitment and activity of myeloid and T cells can be impacted by p53 loss or mutation in malignancies, allowing immune evasion and accelerating cancer growth. Additionally, p53 can work in immune cells, which can have various effects that either hinder or assist the growth of tumors. In this review article, we examined different mutations of P53 in some significant cancers, such as liver, colorectal, and prostate, and reviewed some new therapeutic approaches.
Collapse
Affiliation(s)
- Navid Shomali
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Amin Kamrani
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Nasiri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Ahmadian Heris
- Department of Allergy and Clinical Immunology, Pediatric Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parviz Shahabi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Mohammadinasab
- Department of History of Medicine, School of Traditional Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahram Sadeghvand
- Pediatrics Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Morteza Akbari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
7
|
Gaur T, Poddutoori R, Khare L, Bagal B, Rashmi S, Patkar N, Tembhare P, Pg S, Shetty D, Dutt A, Zhang Q, Konopleva M, Platzbeckar U, Gupta S, Samajdar S, Ramchandra M, Khattry N, Hasan SK. Novel covalent CDK7 inhibitor potently induces apoptosis in acute myeloid leukemia and synergizes with Venetoclax. J Exp Clin Cancer Res 2023; 42:186. [PMID: 37507802 PMCID: PMC10386772 DOI: 10.1186/s13046-023-02750-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
INTRODUCTION The emergence of resistance to the highly successful BCL2-directed therapy is a major unmet need in acute myeloid leukemia (AML), an aggressive malignancy with poor survival rates. Towards identifying therapeutic options for AML patients who progress on BCL2-directed therapy, we studied a clinical-stage CDK7 inhibitor XL102, which is being evaluated in solid tumors (NCT04726332). MATERIALS AND METHODS To determine the anti-proliferative effects of XL102, we performed experiments including time-resolved fluorescence resonance energy transfer, target occupancy, cell cycle and apoptosis-based assays. We also included genetically characterized primary myeloid blasts from de novo and relapsed/refractory AML patients. For mechanistic studies, CRISPR/Cas9 mediated knockout of CDK7 and c-Myc and immunoblotting were performed. NOD/SCID orthotropic and subcutaneous AML xenografts were used to determine anti-leukemic effects. To assess the synergistic effects of XL102 with Venetoclax, we performed RNA sequencing and gene set enrichment analysis using Venetoclax sensitive and resistant model systems. RESULTS XL102, a highly specific, orally bioavailable covalent inhibitor of CDK7. Inhibitory effect on CDK7 by XL102 in primary myeloid blasts (n = 54) was in nanomolar range (mean = 300 nM; range = 4.0-952 nM). XL102 treated AML cells showed a reduction in phosphorylation levels of Serine 2/5/7 at carboxy-terminal domain of RNA polymerase II. T-loop phosphorylation of CDK1(Thr161) and CDK2(Thr160) was inhibited by XL102 in dose-dependent manner leading to cell-cycle arrest. c-Myc downregulation and enhanced levels of p53 and p21 in XL102 treated cells were observed. Increased levels of p21 and activation of p53 by XL102 were mimicked by genetic ablation of CDK7, which supports that the observed effects of XL102 are due to CDK7 inhibition. XL102 treated AML xenografts showed remarkable reduction in hCD45 + marrow cells (mean = 0.60%; range = 0.04%-3.53%) compared to vehicle control (mean = 38.2%; range = 10.1%-78%), with corresponding increase in p53, p21 and decrease in c-Myc levels. The data suggests XL102 induces apoptosis in AML cells via CDK7/c-Myc/p53 axis. RNA-sequencing from paired Venetoclax-sensitive and Venetoclax-resistant cells treated with XL102 showed downregulation of genes involved in proliferation and apoptosis. CONCLUSION Taken together, XL102 with Venetoclax led to synergistic effects in overcoming resistance and provided a strong rationale for clinical evaluation of XL102 as a single agent and in combination with Venetoclax.
Collapse
Affiliation(s)
- Tarang Gaur
- Hasan Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi, Mumbai, 410210, India
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India
| | - Ramulu Poddutoori
- Aurigene Oncology Limited, Electronic City Hosur Road, Bangalore, 560100, India
| | - Leena Khare
- Aurigene Oncology Limited, Electronic City Hosur Road, Bangalore, 560100, India
| | - Bhausaheb Bagal
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, 400014, India
| | - Sonal Rashmi
- Dutt Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi, Mumbai, 410210, India
- Present Address: CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Nikhil Patkar
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India
- Hematopathology Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi, Mumbai, 410210, India
| | - Prashant Tembhare
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India
- Hematopathology Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi, Mumbai, 410210, India
| | - Subramanian Pg
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India
- Hematopathology Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi, Mumbai, 410210, India
| | - Dhanlaxmi Shetty
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India
- Department of Cytogenetics, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi, Mumbai, 410210, India
| | - Amit Dutt
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India
- Dutt Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi, Mumbai, 410210, India
| | - Qi Zhang
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | | | - Uwe Platzbeckar
- Medical Clinic and Policlinic I, Hematology and Cellular Therapy, University Hospital Leipzig, Johannisallee 32, 04103, Leipzig, Germany
| | - Sudeep Gupta
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, 400014, India
| | - Susanta Samajdar
- Aurigene Oncology Limited, Electronic City Hosur Road, Bangalore, 560100, India
| | - Murali Ramchandra
- Aurigene Oncology Limited, Electronic City Hosur Road, Bangalore, 560100, India
| | - Navin Khattry
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India.
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Mumbai, 400014, India.
| | - Syed K Hasan
- Hasan Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi, Mumbai, 410210, India.
- Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, 400094, India.
| |
Collapse
|
8
|
Pei HZ, Peng Z, Zhuang X, Wang X, Lu B, Guo Y, Zhao Y, Zhang D, Xiao Y, Gao T, Yu L, He C, Wu S, Baek SH, Zhao ZJ, Xu X, Chen Y. miR-221/222 induce instability of p53 By downregulating deubiquitinase YOD1 in acute myeloid leukemia. Cell Death Discov 2023; 9:249. [PMID: 37454155 DOI: 10.1038/s41420-023-01537-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/20/2023] [Accepted: 06/28/2023] [Indexed: 07/18/2023] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy characterized by the impaired differentiation and uncontrolled proliferation of myeloid blasts. Tumor suppressor p53 is often downregulated in AML cells via ubiquitination-mediated degradation. While the role of E3 ligase MDM2 in p53 ubiquitination is well-accepted, little is known about the involvement of deubiquitinases (DUBs). Herein, we found that the expression of YOD1, among several DUBs, is substantially reduced in blood cells from AML patients. We identified that YOD1 deubiqutinated and stabilized p53 through interaction via N-terminus of p53 and OTU domain of YOD1. In addition, expression levels of YOD1 were suppressed by elevated miR-221/222 in AML cells through binding to the 3' untranslated region of YOD1, as verified by reporter gene assays. Treatment of cells with miR-221/222 mimics and inhibitors yielded the expected effects on YOD1 expressions, in agreement with the negative correlation observed between the expression levels of miR-221/222 and YOD1 in AML cells. Finally, overexpression of YOD1 stabilized p53, upregulated pro-apoptotic p53 downstream genes, and increased the sensitivity of AML cells to FLT3 inhibitors remarkably. Collectively, our study identified a pathway connecting miR-221/222, YOD1, and p53 in AML. Targeting miR-221/222 and stimulating YOD1 activity may improve the therapeutic effects of FLT3 inhibitors in patients with AML.
Collapse
Affiliation(s)
- Han Zhong Pei
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Zhiyong Peng
- Nanfang-Chunfu Children's Institute of Hematology, Taixin Hospital, Dongguan, Guangdong, China
| | - Xiaomei Zhuang
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Xiaobo Wang
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Bo Lu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Yao Guo
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Yuming Zhao
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Dengyang Zhang
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Yunjun Xiao
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Tianshun Gao
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Liuting Yu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Chunxiao He
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Shunjie Wu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Suk-Hwan Baek
- Department of Biochemistry & Molecular Biology, College of Medicine, Yeungnam University, 170 Hyeonchung-ro, Nam-gu, Daegu, 42415, South Korea.
| | - Zhizhuang Joe Zhao
- Department of Pathology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd., BMSB 451, Oklahoma City, OK, 73104, USA.
| | - Xiaojun Xu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China.
| | - Yun Chen
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong, China.
| |
Collapse
|
9
|
Pourebrahim R, Heinz Montoya R, Alaniz Z, Ostermann L, Lin PP, Liu B, Ayoub E, Burks JK, Andreeff M. Mdm2/p53 levels in bone marrow mesenchymal stromal cells are essential for maintaining the hematopoietic niche in response to DNA damage. Cell Death Dis 2023; 14:371. [PMID: 37353528 PMCID: PMC10290070 DOI: 10.1038/s41419-023-05844-7] [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: 02/13/2023] [Revised: 04/24/2023] [Accepted: 04/28/2023] [Indexed: 06/25/2023]
Abstract
Mesenchymal stromal cells (MSCs) are a key component of the bone marrow (BM) niche, providing essential support required for the maintenance of hematopoietic stem cells. To advance our understanding of physiological functions of p53 and Mdm2 in BM-MSCs, we developed traceable conditional mouse models targeting Mdm2 and/or Trp53 in vivo. We demonstrate that Mdm2 is essential for the emergence, maintenance, and hematopoietic support of BM-MSCs. Mdm2 haploinsufficiency in BM-MSCs resulted in genotoxic stress-associated thrombocytopenia, suggesting a functional role for Mdm2 in hematopoiesis. In a syngeneic mouse model of acute myeloid leukemia (AML), Trp53 deletion in BM-MSCs improved survival, and protected BM against hematopoietic toxicity from a murine Mdm2i, DS-5272. The transcriptional changes were associated with dysregulation of glycolysis, gluconeogenesis, and Hif-1α in BM-MSCs. Our results reveal a physiologic function of Mdm2 in BM-MSC, identify a previously unknown role of p53 pathway in BM-MSC-mediated support in AML and expand our understanding of the mechanism of hematopoietic toxicity of MDM2is.
Collapse
Affiliation(s)
- Rasoul Pourebrahim
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rafael Heinz Montoya
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zoe Alaniz
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lauren Ostermann
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Patrick P Lin
- Department of Orthopedic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bin Liu
- Department of Epigenetic and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Edward Ayoub
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jared K Burks
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
10
|
Zanjirband M, Rahgozar S, Aberuyi N. miR-16-5p enhances sensitivity to RG7388 through targeting PPM1D expression (WIP1) in Childhood Acute Lymphoblastic Leukemia. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:242-256. [PMID: 37457129 PMCID: PMC10344722 DOI: 10.20517/cdr.2022.113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 03/02/2023] [Accepted: 04/28/2023] [Indexed: 07/18/2023]
Abstract
Aim: Given the encouraging results of the p53-Mdm2 inhibitor RG7388 in clinical trials and the vital function of miR-16-5p in suppressing cell proliferation, the aim of the present study was to investigate the combined impact of RG7388 and miR-16-5p overexpression on the childhood acute lymphoblastic leukemia (chALL). Methods: miRTarBase and miRDB, along with KEGG and STRING databases, were used to predict miR-16-5p target genes and explore protein-protein interaction networks, respectively. B- and T-lymphoblastic cell lines, in addition to patient primary cells, were treated with RG7388. Ectopic overexpression of miR-16-5p in Nalm6 cell line was induced through cell electroporation and transfection of microRNA mimics was confirmed by qRT-PCR. Cell viability was evaluated using the MTT assay. Western blot analyses were performed to evaluate the effects of RG7388 and miR-16-5p upregulation on the protein levels of p53 and its downstream target genes in chALL cells. Paired sample t-test was employed for statistical analyses. Results: MTT assay showed RG7388-induced cytotoxicity in wild-type p53 Nalm6 cell line and p53 functional patient primary cells. However, CCRF-CEM and p53 non-functional leukemic cells indicated drug resistance. Western blot analyses validated the bioinformatics results, confirming the downregulation of WIP1, p53 stabilization, as well as overexpression of p21WAF1 and Mdm2 proteins in Nalm6 cells transfected with miR-16-5p. Moreover, enhanced sensitivity to RG7388 was observed in the transfected cells. Conclusion: This is the first study indicating the mechanistic importance of miR-16-5p overexpression in chALL and its inhibitory role in leukemia treatment when combined with the p53-Mdm2 antagonist, RG7388. These findings might be useful for researchers and clinicians to pave the way for better management of chALL.
Collapse
Affiliation(s)
- Maryam Zanjirband
- Correspondence to: Dr. Soheila Rahgozar, Dr. Maryam Zanjirband, Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Hezar Jerib Avenue, Isfahan 15100, Iran. E-mail: ;
| | - Soheila Rahgozar
- Correspondence to: Dr. Soheila Rahgozar, Dr. Maryam Zanjirband, Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Hezar Jerib Avenue, Isfahan 15100, Iran. E-mail: ;
| | | |
Collapse
|
11
|
Pourebrahim R, Montoya RH, Alaniz Z, Ostermann L, Lin PP, Liu B, Ayoub E, Burks JK, Andreeff M. Mdm2/p53 levels in bone marrow mesenchymal stromal cells is essential for maintaining the hematopoietic niche in response to DNA damage. RESEARCH SQUARE 2023:rs.3.rs-2544760. [PMID: 36909480 PMCID: PMC10002809 DOI: 10.21203/rs.3.rs-2544760/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Mesenchymal stromal cells (MSCs) are a key component of the bone marrow (BM) niche, providing essential support required for maintenance of hematopoietic stem cells. To advance our understanding of physiological functions of p53 and Mdm2 in BM-MSCs, we developed traceable conditional mouse models targeting Mdm2 and/or Trp53 in vivo . We demonstrate that Mdm2 is essential for the emergence, maintenance and hematopoietic support of BM-MSCs. Mdm2 haploinsufficiency in BM-MSCs resulted in genotoxic stress-associated thrombocytopenia, suggesting a functional role for Mdm2 in hematopoiesis. In a syngeneic mouse model of acute myeloid leukemia (AML), Trp53 deletion in BM-MSCs improved survival, and protected BM against hematopoietic toxicity from a murine Mdm2i, DS-5272. The transcriptional changes were associated with dysregulation of glycolysis, gluconeogenesis, and Hif-1α in BM-MSCs. Our results reveal a physiologic function of Mdm2 in BM-MSC, identify a previously unknown role of p53 pathway in BM-MSC-mediated support in AML and expand our understanding of the mechanism of hematopoietic toxicity of MDM2is.
Collapse
Affiliation(s)
| | | | | | | | | | - Bin Liu
- Epigenetic and Molecular Carcinogenesis
| | | | | | | |
Collapse
|
12
|
Tripathi R, Anifowose A, Lu W, Yang X, Wang B. Upregulation of p53 through induction of MDM2 degradation: improved potency through the introduction of an alkylketone sidechain on the anthraquinone core. J Enzyme Inhib Med Chem 2022; 37:2370-2381. [PMID: 36043494 PMCID: PMC9448394 DOI: 10.1080/14756366.2022.2116699] [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] [Indexed: 11/22/2022] Open
Abstract
Overexpression of ubiquitin ligase MDM2 causes depletion of the p53 tumour-suppressor and thus leads to cancer progression. In recent years, anthraquinone analogs have received significant attention due to their ability to downregulate MDM2, thereby promoting p53-induced apoptosis. Previously, we have developed potent anthraquinone compounds having the ability to upregulate p53 via inhibition of MDM2 in both cell culture and animal models of acute lymphocytic leukaemia. Earlier work was focussed on mechanistic work, pharmacological validation of this class of compounds in animal models, and mapping out structural space that allows for further modification and optimisation. Herein, we describe our work in optimising the substituents on the two phenol hydroxyl groups. It was found that the introduction of an alkylketone moiety led to a potent series of analogs with BW-AQ-350 being the most potent compound yet (IC50 = 0.19 ± 0.01 µM) which exerts cytotoxicity by inducing MDM2 degradation and p53 upregulation.
Collapse
Affiliation(s)
- Ravi Tripathi
- Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Abiodun Anifowose
- Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Wen Lu
- Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Xiaoxiao Yang
- Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| | - Binghe Wang
- Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
13
|
Chang M, Gao F, Chen J, Gnawali G, Wang W. MDM2-BCL-X L PROTACs enable degradation of BCL-X L and stabilization of p53. ACTA MATERIA MEDICA 2022; 1:333-342. [PMID: 36910255 PMCID: PMC10004178 DOI: 10.15212/amm-2022-0022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Inhibition or degradation of anti-apoptotic protein BCL-XL is a viable strategy for cancer treatment. Despite the recent development of PROTACs for degradation of BCL-XL, the E3 ligases are confined to the commonly used VHL and CRBN. Herein we report the development of MDM2-BCL-XL PROTACs using MDM2 as E3 ligase for degradation of BCL-XL. Three MDM2-BCL-XL PROTACs derived from MDM2 inhibitor Nutlin-3, which can also upregulate p53, and BCL-2/BCL-XL inhibitor ABT-263 with different linker length were designed, synthesized, and evaluated in vitro. We found BMM4 exhibited potent, selective degradation activity against BCL-XL and stabilized tumor suppressor p53 in U87, A549 and MV-4-11 cancer cell lines. Moreover, combination of BMM4 and BCL-2 inhibitor ABT-199 showed synergistic antiproliferative activity. The unique dual-functional PROTACs offers an alternative strategy for targeted protein degradation.
Collapse
Affiliation(s)
- Mengyang Chang
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, USA
| | - Feng Gao
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Az, USA
| | - Jing Chen
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Az, USA
| | - Giri Gnawali
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Az, USA
| | - Wei Wang
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, USA.,Department of Pharmacology and Toxicology, University of Arizona, Tucson, Az, USA
| |
Collapse
|
14
|
AMG-232, a New Inhibitor of MDM-2, Enhance Doxorubicin Efficiency in Pre-B Acute Lymphoblastic Leukemia Cells. Rep Biochem Mol Biol 2022; 11:111-124. [PMID: 35765530 PMCID: PMC9208559 DOI: 10.52547/rbmb.11.1.111] [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: 01/04/2022] [Accepted: 01/09/2022] [Indexed: 01/11/2023]
Abstract
Background Doxorubicin (DOX)-induced cardiotoxicity appears to be a growing concern for extensive use in acute lymphoblastic leukemia (ALL). The new combination treatment strategies, therefore might be an effective way of decreasing its side effects as well as improving efficacy. AMG232 (KRT-232) is a potential MDM-2 inhibitor, increasing available p53 through disturbing p53-MDM-2 interaction. In this study, we examined the effects of AMG232 on DOX-induced apoptosis of NALM-6 cells. Methods The anti-leukemic effects of Doxorubicin on NALM-6 cells, either alone or in combination with AMG232, were confirmed by MTT assay, Annexin/PI apoptosis assay, and cell cycle analysis. Expression of apoptosis and autophagy-related genes were further evaluated by Real time-PCR method. To investigate the effect of AMG232 on NALM-6 cells, the activation of p53, p21, MDM-2, cleaved Caspase-3 proteins was evaluated using western blot analysis. Results The results showed that AMG232 inhibition of MDM-2 enhances Doxorubicin-induced apoptosis in NALM-6 cells through caspase-3 activation in a time and dose-dependent manner. Furthermore, co-treatment of AMG232 with Doxorubicin hampered the transition of NALM-6 cells from G1 phase through increasing p21 protein. In addition, this combination treatment led to enhanced expression of apoptosis and autophagy-related genes in ALL cell lines. Conclusion The results declared that AMG232 as an MDM-2 inhibitor could be an effective approach to enhance antitumor effects of Doxorubicin on NALM-6 cells as well as an effective future treatment for ALL patients.
Collapse
|
15
|
Combined Drug Targeting of p53-dependent and -independent Pathways Depletes Myelofibrosis Hematopoietic Stem/Progenitor Cells. Leukemia 2022; 36:733-745. [PMID: 34642468 PMCID: PMC8885407 DOI: 10.1038/s41375-021-01446-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/21/2021] [Accepted: 09/29/2021] [Indexed: 01/06/2023]
Abstract
Current therapy for myelofibrosis (MF) results in a limited prolongation of patient survival. In order to improve treatment outcomes, we developed a strategy to effectively deplete MF hematopoietic stem/progenitor cells (HSPCs). In the present study, an imipridone, ONC201, was combined with RG7112, an antagonist of MDM2, a p53 negative regulator, to activate downstream events of the p53 and TNF-related apoptosis-inducing ligand (TRAIL)/death receptor (DR) pathways. As compared to treatment with the individual drugs, the combination of ONC201 and RG7112 promoted greater degrees of apoptosis of MF CD34+ cells through activation of both p53-dependent and -independent pathways. Importantly, treatment with ONC201-RG7112 not only decreased the number of JAK2V617F+ and calreticulin mutated colonies assayed from MF CD34+ cells, but allowed for the persistence or appearance of JAK2 wild type colonies. Treatment with ONC201 combined with RG7112 could be a potentially effective strategy for treating MF patients.
Collapse
|
16
|
Fink A, Hung E, Singh I, Ben-Neriah Y. Immunity in acute myeloid leukemia: Where the immune response and targeted therapy meet. Eur J Immunol 2021; 52:34-43. [PMID: 34648664 DOI: 10.1002/eji.202048945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 07/29/2021] [Accepted: 10/05/2021] [Indexed: 12/19/2022]
Abstract
Acute myeloid leukemia (AML) is a highly aggressive disease with high relapse and mortality rates. Recent years have shown a surge in novel therapeutic development for AML, both in clinical and preclinical stages. These developments include targeted therapies based on AML-specific molecular signatures as well as more general immune modulation and vaccination studies. In this review, we will explore the evolving arena of AML therapy and suggest some intriguing connections between immune system modulation and targeted therapy. Improved understanding of the immune system involvement in various stages of the disease and the crosstalk between immune effectors, targeted therapy, and AML cells can provide a better framework for designing the next generation of AML therapies.
Collapse
Affiliation(s)
- Avner Fink
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Eric Hung
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Indranil Singh
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Yinon Ben-Neriah
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
17
|
Haines E, Nishida Y, Carr MI, Montoya RH, Ostermann LB, Zhang W, Zenke FT, Blaukat A, Andreeff M, Vassilev LT. DNA-PK inhibitor peposertib enhances p53-dependent cytotoxicity of DNA double-strand break inducing therapy in acute leukemia. Sci Rep 2021; 11:12148. [PMID: 34108527 PMCID: PMC8190296 DOI: 10.1038/s41598-021-90500-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 05/10/2021] [Indexed: 12/14/2022] Open
Abstract
Peposertib (M3814) is a potent and selective DNA-PK inhibitor in early clinical development. It effectively blocks non-homologous end-joining repair of DNA double-strand breaks (DSB) and strongly potentiates the antitumor effect of ionizing radiation (IR) and topoisomerase II inhibitors. By suppressing DNA-PK catalytic activity in the presence of DNA DSB, M3814 potentiates ATM/p53 signaling leading to enhanced p53-dependent antitumor activity in tumor cells. Here, we investigated the therapeutic potential of M3814 in combination with DSB-inducing agents in leukemia cells and a patient-derived tumor. We show that in the presence of IR or topoisomerase II inhibitors, M3814 boosts the ATM/p53 response in acute leukemia cells leading to the elevation of p53 protein levels as well as its transcriptional activity. M3814 synergistically sensitized p53 wild-type, but not p53-deficient, AML cells to killing by DSB-inducing agents via p53-dependent apoptosis involving both intrinsic and extrinsic effector pathways. The antileukemic effect was further potentiated by enhancing daunorubicin-induced myeloid cell differentiation. Further, combined with the fixed-ratio liposomal formulation of daunorubicin and cytarabine, CPX-351, M3814 enhanced the efficacy against leukemia cells in vitro and in vivo without increasing hematopoietic toxicity, suggesting that DNA-PK inhibition could offer a novel clinical strategy for harnessing the anticancer potential of p53 in AML therapy.
Collapse
MESH Headings
- Animals
- Apoptosis
- Cell Proliferation
- DNA Breaks, Double-Stranded
- DNA Repair
- DNA-Activated Protein Kinase/antagonists & inhibitors
- Gene Expression Regulation, Leukemic
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Male
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Phosphorylation
- Protein Kinase Inhibitors/pharmacology
- Pyridazines/pharmacology
- Quinazolines/pharmacology
- Signal Transduction
- Tumor Cells, Cultured
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Eric Haines
- Translational Innovation Platform Oncology and Immuno-Oncology, EMD Serono Research & Development Institute, Inc, Billerica, MA, USA
| | - Yuki Nishida
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael I Carr
- Translational Innovation Platform Oncology and Immuno-Oncology, EMD Serono Research & Development Institute, Inc, Billerica, MA, USA
| | - Rafael Heinz Montoya
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lauren B Ostermann
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Weiguo Zhang
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Frank T Zenke
- Translational Innovation Platform Oncology and Immuno-Oncology, Merck KGaA, Darmstadt, Germany
| | - Andree Blaukat
- Translational Innovation Platform Oncology and Immuno-Oncology, Merck KGaA, Darmstadt, Germany
| | - Michael Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| | - Lyubomir T Vassilev
- Translational Innovation Platform Oncology and Immuno-Oncology, EMD Serono Research & Development Institute, Inc, Billerica, MA, USA.
| |
Collapse
|
18
|
Achieving effective and selective CK1 inhibitors through structure modification. Future Med Chem 2021; 13:505-528. [PMID: 33438471 DOI: 10.4155/fmc-2020-0215] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Casein kinase 1 (CK1) is an extensively expressed serine/threonine kinase family, with six highly conserved isoforms of human CK1. Due to its involvement in many biological processes, CK1 is a promising target for several pathological states, including circadian sleep disorder, neurodegenerative diseases, cancer and inflammation. However, due to the structural similarities between the six CK1 members, the design of CK1 inhibitors is intricate. So far, no effective CK1 inhibitors are reported to reach clinical trials; thus, approaches to obtaining both selective and effective CK1 inhibitors are in great demand. Here we analyze several CK1 inhibitors that provide successful experience for structure-based drug design and rational structure modification, which could provide references for further drug design.
Collapse
|
19
|
Kannan S, Aronica PGA, Ng S, Gek Lian DT, Frosi Y, Chee S, Shimin J, Yuen TY, Sadruddin A, Kaan HYK, Chandramohan A, Wong JH, Tan YS, Chang ZW, Ferrer-Gago FJ, Arumugam P, Han Y, Chen S, Rénia L, Brown CJ, Johannes CW, Henry B, Lane DP, Sawyer TK, Verma CS, Partridge AW. Macrocyclization of an all-d linear α-helical peptide imparts cellular permeability. Chem Sci 2020; 11:5577-5591. [PMID: 32874502 PMCID: PMC7441689 DOI: 10.1039/c9sc06383h] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 05/08/2020] [Indexed: 12/13/2022] Open
Abstract
Peptide-based molecules hold great potential as targeted inhibitors of intracellular protein-protein interactions (PPIs). Indeed, the vast diversity of chemical space conferred through their primary, secondary and tertiary structures allows these molecules to be applied to targets that are typically deemed intractable via small molecules. However, the development of peptide therapeutics has been hindered by their limited conformational stability, proteolytic sensitivity and cell permeability. Several contemporary peptide design strategies are aimed at addressing these issues. Strategic macrocyclization through optimally placed chemical braces such as olefinic hydrocarbon crosslinks, commonly referred to as staples, may improve peptide properties by (i) restricting conformational freedom to improve target affinities, (ii) improving proteolytic resistance, and (iii) enhancing cell permeability. As a second strategy, molecules constructed entirely from d-amino acids are hyper-resistant to proteolytic cleavage, but generally lack conformational stability and membrane permeability. Since neither approach is a complete solution, we have combined these strategies to identify the first examples of all-d α-helical stapled and stitched peptides. As a template, we used a recently reported all d-linear peptide that is a potent inhibitor of the p53-Mdm2 interaction, but is devoid of cellular activity. To design both stapled and stitched all-d-peptide analogues, we used computational modelling to predict optimal staple placement. The resultant novel macrocyclic all d-peptide was determined to exhibit increased α-helicity, improved target binding, complete proteolytic stability and, most notably, cellular activity.
Collapse
Affiliation(s)
- Srinivasaraghavan Kannan
- Bioinformatics Institute , Agency for Science, Technology and Research (ASTAR) , 30 Biopolis Street, #07-01 Matrix , Singapore 138671 , Singapore . ; ; ; Tel: +65 6478 8353 ; Tel: +65 6478 8273
| | - Pietro G A Aronica
- Bioinformatics Institute , Agency for Science, Technology and Research (ASTAR) , 30 Biopolis Street, #07-01 Matrix , Singapore 138671 , Singapore . ; ; ; Tel: +65 6478 8353 ; Tel: +65 6478 8273
| | - Simon Ng
- p53 Laboratory , Agency for Science, Technology and Research (ASTAR) , 8A Biomedical Grove, #06-04/05, Neuros/Immunos , Singapore 138648
| | - Dawn Thean Gek Lian
- p53 Laboratory , Agency for Science, Technology and Research (ASTAR) , 8A Biomedical Grove, #06-04/05, Neuros/Immunos , Singapore 138648
| | - Yuri Frosi
- p53 Laboratory , Agency for Science, Technology and Research (ASTAR) , 8A Biomedical Grove, #06-04/05, Neuros/Immunos , Singapore 138648
| | - Sharon Chee
- p53 Laboratory , Agency for Science, Technology and Research (ASTAR) , 8A Biomedical Grove, #06-04/05, Neuros/Immunos , Singapore 138648
| | - Jiang Shimin
- p53 Laboratory , Agency for Science, Technology and Research (ASTAR) , 8A Biomedical Grove, #06-04/05, Neuros/Immunos , Singapore 138648
| | - Tsz Ying Yuen
- Institute of Chemical & Engineering Science , Agency for Science, Technology and Research (ASTAR) , 8 Biomedical Grove, #07, Neuros Building , Singapore 138665
| | - Ahmad Sadruddin
- MSD International , Translation Medicine Research Centre , 8 Biomedical Grove, #04-01/05 Neuros Building , Singapore , 138665 , Singapore .
| | - Hung Yi Kristal Kaan
- MSD International , Translation Medicine Research Centre , 8 Biomedical Grove, #04-01/05 Neuros Building , Singapore , 138665 , Singapore .
| | - Arun Chandramohan
- MSD International , Translation Medicine Research Centre , 8 Biomedical Grove, #04-01/05 Neuros Building , Singapore , 138665 , Singapore .
| | - Jin Huei Wong
- Bioinformatics Institute , Agency for Science, Technology and Research (ASTAR) , 30 Biopolis Street, #07-01 Matrix , Singapore 138671 , Singapore . ; ; ; Tel: +65 6478 8353 ; Tel: +65 6478 8273
| | - Yaw Sing Tan
- Bioinformatics Institute , Agency for Science, Technology and Research (ASTAR) , 30 Biopolis Street, #07-01 Matrix , Singapore 138671 , Singapore . ; ; ; Tel: +65 6478 8353 ; Tel: +65 6478 8273
| | - Zi Wei Chang
- Singapore Immunology Network (SIgN) , Agency for Science, Technology and Research (ASTAR) , 8A Biomedical Grove, #03-06, Immunos , Singapore 138648
| | - Fernando J Ferrer-Gago
- p53 Laboratory , Agency for Science, Technology and Research (ASTAR) , 8A Biomedical Grove, #06-04/05, Neuros/Immunos , Singapore 138648
| | - Prakash Arumugam
- Bioinformatics Institute , Agency for Science, Technology and Research (ASTAR) , 30 Biopolis Street, #07-01 Matrix , Singapore 138671 , Singapore . ; ; ; Tel: +65 6478 8353 ; Tel: +65 6478 8273
| | - Yi Han
- Merck & Co., Inc. , Kenilworth , New Jersey , USA
| | - Shiying Chen
- Merck & Co., Inc. , Kenilworth , New Jersey , USA
| | - Laurent Rénia
- Singapore Immunology Network (SIgN) , Agency for Science, Technology and Research (ASTAR) , 8A Biomedical Grove, #03-06, Immunos , Singapore 138648
| | - Christopher J Brown
- p53 Laboratory , Agency for Science, Technology and Research (ASTAR) , 8A Biomedical Grove, #06-04/05, Neuros/Immunos , Singapore 138648
| | - Charles W Johannes
- Institute of Chemical & Engineering Science , Agency for Science, Technology and Research (ASTAR) , 8 Biomedical Grove, #07, Neuros Building , Singapore 138665
| | - Brian Henry
- MSD International , Translation Medicine Research Centre , 8 Biomedical Grove, #04-01/05 Neuros Building , Singapore , 138665 , Singapore .
| | - David P Lane
- p53 Laboratory , Agency for Science, Technology and Research (ASTAR) , 8A Biomedical Grove, #06-04/05, Neuros/Immunos , Singapore 138648
| | | | - Chandra S Verma
- Bioinformatics Institute , Agency for Science, Technology and Research (ASTAR) , 30 Biopolis Street, #07-01 Matrix , Singapore 138671 , Singapore . ; ; ; Tel: +65 6478 8353 ; Tel: +65 6478 8273
- School of Biological Sciences , Nanyang Technological University , 60 Nanyang Drive , Singapore 637551
- Department of Biological Sciences , National University of Singapore , 14 Science Drive 4 , Singapore 117543
| | - Anthony W Partridge
- MSD International , Translation Medicine Research Centre , 8 Biomedical Grove, #04-01/05 Neuros Building , Singapore , 138665 , Singapore .
| |
Collapse
|
20
|
Carr MI, Zimmermann A, Chiu LY, Zenke FT, Blaukat A, Vassilev LT. DNA-PK Inhibitor, M3814, as a New Combination Partner of Mylotarg in the Treatment of Acute Myeloid Leukemia. Front Oncol 2020; 10:127. [PMID: 32117773 PMCID: PMC7031257 DOI: 10.3389/fonc.2020.00127] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/24/2020] [Indexed: 02/03/2023] Open
Abstract
Despite significant advances in the treatment of acute myeloid leukemia (AML) the long-term prognosis remains relatively poor and there is an urgent need for improved therapies with increased potency and tumor selectivity. Mylotarg is the first AML-targeting drug from a new generation of antibody drug conjugate (ADC) therapies aiming at the acute leukemia cell compartment with increased specificity. This agent targets leukemia cells for apoptosis with a cytotoxic payload, calicheamicin, carried by a CD33-specific antibody. Calicheamicin induces DNA double strand breaks (DSB) which, if left unrepaired, lead to cell cycle arrest and apoptosis in cancer cells. However, repair of DSB by the non-homologous end joining pathway driven by DNA-dependent protein kinase (DNA-PK) can reduce the efficacy of calicheamicin. M3814 is a novel, potent and selective inhibitor of DNA-PK. This compound effectively blocks DSB repair, strongly potentiates the antitumor activity of ionizing radiation and DSB-inducing chemotherapeutics and is currently under clinical investigation. Suppressing DSB repair with M3814 synergistically enhanced the apoptotic activity of calicheamicin in cultured AML cells. Combination of M3814 with Mylotarg in two AML xenograft models, MV4-11 and HL-60, demonstrated increased efficacy and significantly improved survival benefit without elevated body weight loss. Our results support a new application for pharmacological DNA-PK inhibitors as enhancers of Mylotarg and a potential new combination treatment option for AML patients.
Collapse
Affiliation(s)
- Michael I. Carr
- Translational Innovation Platform Oncology, EMD Serono Research and Development Institute, Inc., Billerica, MA, United States
| | - Astrid Zimmermann
- Translational Innovation Platform Oncology, Merck KGaA, Darmstadt, Germany
| | - Li-Ya Chiu
- Translational Innovation Platform Oncology, EMD Serono Research and Development Institute, Inc., Billerica, MA, United States
| | - Frank T. Zenke
- Translational Innovation Platform Oncology, Merck KGaA, Darmstadt, Germany
| | - Andree Blaukat
- Translational Innovation Platform Oncology, Merck KGaA, Darmstadt, Germany
| | - Lyubomir T. Vassilev
- Translational Innovation Platform Oncology, EMD Serono Research and Development Institute, Inc., Billerica, MA, United States
| |
Collapse
|
21
|
Rationale for a Combination Therapy Consisting of MCL1- and MEK-Inhibitors in Acute Myeloid Leukemia. Cancers (Basel) 2019; 11:cancers11111779. [PMID: 31718075 PMCID: PMC6896073 DOI: 10.3390/cancers11111779] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/07/2019] [Accepted: 11/08/2019] [Indexed: 11/17/2022] Open
Abstract
Amplification and overexpression of the myeloid cell leukemia differentiation protein MCL1 and the murine double minute protein MDM2 have been reported in various human tumors as well as hematological malignancies including acute myeloid leukemia (AML). While MCL1 is an anti-apoptotic member of the BCL-2 family proteins, MDM2 is an important cellular inhibitor of the p53 tumor suppressor. The key oncogene in AML is the FLT3 growth factor receptor gene. FLT3 signaling pathways including the MAPK cascade (RAS-RAF-MEK-ERK) are highly active in AML cells, leading to induced protein translation and cell proliferation as well as reduced apoptosis. Consequently, combined administration of MCL1-, MDM2-, and MEK-inhibitors may present a promising anti-leukemic treatment strategy. Here, we assessed the MCL1-antagonist S63845, the MDM2-inhibitor HDM201, and the MEK1/2-inhibitor trametinib as single agents and in combination in a variety of AML cell lines and mononuclear cells isolated from patients with hematological malignancies centered on myeloid leukemia, some lymphatic leukemia, as well as some lymphomas, for their ability to induce apoptosis and cell death. We observed a considerably varying anti-leukemic efficacy of the MCL1-inhibitor S63845 and the MEK1/2-inhibitor trametinib. Hematological cells with susceptibility to the single compounds as well as to the combined treatment were defined by elevated MCL1- and MEK-protein levels, independent of the mutational status of FLT3 and TP53. Our data indicate that hematological cells with elevated MCL1- and MEK-protein levels are most sensitive to the combined treatment with S63845 and trametinib. MCL1- and MEK1/2-protein expression may be valid biomarkers for treatment response to S63845 and trametinib, respectively.
Collapse
|
22
|
Bissinger R, Lang E, Gonzalez-Menendez I, Quintanilla-Martinez L, Ghashghaeinia M, Pelzl L, Sukkar B, Bhuyan AAM, Salker MS, Singh Y, Fehrenbacher B, Fakhri H, Umbach AT, Schaller M, Qadri SM, Lang F. Genetic deficiency of the tumor suppressor protein p53 influences erythrocyte survival. Apoptosis 2019; 23:641-650. [PMID: 30238335 DOI: 10.1007/s10495-018-1481-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The transcription factor p53 suppresses tumor growth by inducing nucleated cell apoptosis and cycle arrest. Because of its influence on primitive erythroid cell differentiation and survival, p53 is an important determinant of erythropoiesis. However, the impact of p53 on the fate of erythrocytes, cells lacking nucleus and mitochondria, during their post-maturation phase in the circulation remained elusive. Erythrocyte survival may be compromised by suicidal erythrocyte death or eryptosis, which is hallmarked by phosphatidylserine translocation and stimulated by increase of cytosolic Ca2+ concentration. Here, we comparatively examined erythrocyte homeostasis in p53-mutant mice (Trp53tm1Tyj/J) and in corresponding WT mice (C57BL/6J) by analyzing eryptosis and erythropoiesis. To this end, spontaneous cell membrane phosphatidylserine exposure and cytosolic Ca2+ concentration were higher in erythrocytes drawn from Trp53tm1Tyj/J mice than from WT mice. Eryptosis induced by glucose deprivation, a pathophysiological cell stressor, was slightly, but significantly more prominent in erythrocytes drawn from Trp53tm1Tyj/J mice as compared to WT mice. The loss of erythrocytes by eryptosis was fully compensated by enhanced erythropoiesis in Trp53tm1Tyj/J mice, as reflected by increased reticulocytosis and abundance of erythroid precursor cells in the bone marrow. Accordingly, erythrocyte number, packed cell volume and hemoglobin were similar in Trp53tm1Tyj/J and WT mice. Taken together, functional p53 deficiency enhances the turnover of circulating erythrocytes by parallel increase of eryptosis and stimulated compensatory erythropoiesis.
Collapse
Affiliation(s)
- Rosi Bissinger
- Department of Internal Medicine III, Eberhard-Karls University Tübingen, Tübingen, Germany
| | - Elisabeth Lang
- Department of Molecular Medicine II, Heinrich-Heine University, Düsseldorf, Germany
| | - Irene Gonzalez-Menendez
- Institute of Pathology and Neuropathology, Eberhard-Karls University Tübingen, Tübingen, Germany.,Comprehensive Cancer Center, University Hospital Tübingen, Tübingen, Germany
| | - Leticia Quintanilla-Martinez
- Institute of Pathology and Neuropathology, Eberhard-Karls University Tübingen, Tübingen, Germany.,Comprehensive Cancer Center, University Hospital Tübingen, Tübingen, Germany
| | - Mehrdad Ghashghaeinia
- Psoriasis Center, Department of Dermatology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, Germany.,Department of Vegetative & Clinical Physiology, Institute of Physiology, Eberhard Karls University Tübingen, Wilhelmstraße 56, 72074, Tübingen, Germany
| | - Lisann Pelzl
- Department of Vegetative & Clinical Physiology, Institute of Physiology, Eberhard Karls University Tübingen, Wilhelmstraße 56, 72074, Tübingen, Germany
| | - Basma Sukkar
- Department of Vegetative & Clinical Physiology, Institute of Physiology, Eberhard Karls University Tübingen, Wilhelmstraße 56, 72074, Tübingen, Germany
| | - Abdulla Al Mamun Bhuyan
- Department of Vegetative & Clinical Physiology, Institute of Physiology, Eberhard Karls University Tübingen, Wilhelmstraße 56, 72074, Tübingen, Germany
| | - Madhuri S Salker
- Research Institute for Women's Health, University Hospital Tübingen, Tübingen, Germany
| | - Yogesh Singh
- Institute of Medical Genetics and Applied Genomics, Eberhard-Karls University Tübingen, Tübingen, Germany
| | - Birgit Fehrenbacher
- Department of Dermatology, Eberhard-Karls University Tübingen, Tübingen, Germany
| | - Hajar Fakhri
- Department of Vegetative & Clinical Physiology, Institute of Physiology, Eberhard Karls University Tübingen, Wilhelmstraße 56, 72074, Tübingen, Germany
| | - Anja T Umbach
- Department of Vegetative & Clinical Physiology, Institute of Physiology, Eberhard Karls University Tübingen, Wilhelmstraße 56, 72074, Tübingen, Germany
| | - Martin Schaller
- Department of Dermatology, Eberhard-Karls University Tübingen, Tübingen, Germany
| | - Syed M Qadri
- Department of Pathology and Molecular Medicine, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada. .,Centre for Innovation, Canadian Blood Services, Hamilton, ON, Canada.
| | - Florian Lang
- Department of Internal Medicine III, Eberhard-Karls University Tübingen, Tübingen, Germany. .,Department of Molecular Medicine II, Heinrich-Heine University, Düsseldorf, Germany. .,Department of Vegetative & Clinical Physiology, Institute of Physiology, Eberhard Karls University Tübingen, Wilhelmstraße 56, 72074, Tübingen, Germany.
| |
Collapse
|
23
|
Barbosa K, Li S, Adams PD, Deshpande AJ. The role of TP53 in acute myeloid leukemia: Challenges and opportunities. Genes Chromosomes Cancer 2019; 58:875-888. [DOI: 10.1002/gcc.22796] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/02/2019] [Accepted: 08/05/2019] [Indexed: 12/16/2022] Open
Affiliation(s)
- Karina Barbosa
- Tumor Initiation and Maintenance Program Sanford Burnham Prebys Medical Discovery Institute La Jolla California
| | - Sha Li
- Tumor Initiation and Maintenance Program Sanford Burnham Prebys Medical Discovery Institute La Jolla California
| | - Peter D. Adams
- Tumor Initiation and Maintenance Program Sanford Burnham Prebys Medical Discovery Institute La Jolla California
| | - Aniruddha J. Deshpande
- Tumor Initiation and Maintenance Program Sanford Burnham Prebys Medical Discovery Institute La Jolla California
| |
Collapse
|
24
|
Bajelan B, Zaki-Dizaji M, Rahmani B, Darzi S, Darabi S, Rajaei F. Resistance of human primary mesenchymal stem cells to cytotoxic effects of nutlin-3 in vitro. J Cell Biochem 2019; 121:788-796. [PMID: 31452266 DOI: 10.1002/jcb.29324] [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: 11/11/2018] [Accepted: 01/28/2019] [Indexed: 11/11/2022]
Abstract
BACKGROUND The small-molecule nutlin-3 was found to be an effective therapeutic compound and p53 activator, and acts as a murine double minute 2 antagonist, although these findings need to be clinically confirmed. The essential components of the bone marrow include mesenchymal stem cells (MSCs), which play a key role in protecting, regenerating, and proliferating hematopoietic stem cells (HSCs). This feature is vital for HSC after exposure to myelotoxic anticancer agents; nevertheless, the effects of nutlin-3 on MSCs remain to be disclosed. The present research study was conducted to examine the antiproliferative and proapoptotic effectiveness of nutlin-3 in bone marrow MSCs (BMSCs). MATERIALS AND METHODS Human-derived BMSCs were cultured for different durations, that is, 24, 48, and 72 hours, and treated using various concentrations of nutlin-3, including 5, 10, 25, 50, and 100 μΜ. To investigate the effect of nutlin-3 on the apoptosis, cell vitality and proliferation in BMSCs, the terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), thiazolyl blue tetrazolium bromide, propidium iodide (PI) and annexin V assay, as well as real-time polymerase chain reaction, were used. RESULTS BMSCs viability significantly decreased (P < .05) in the cells treated at concentrations of 50 and 100 μM for 24 hours and concentrations of 25, 50, and 100 μM for 48 hours and at all concentrations for 72 hours. The apoptosis of BMSCs (TUNEL positive) was significantly more visible at concentrations of 25 and 50 μM compared with that in the controls (P < .05), while this increased through dose-dependent processes. Annexin V/PI staining revealed negligible dose-dependent increases in all the apoptotic cells after 72 hours of incubation, and this apoptosis elevation was significant at 25 and 50 μM (P < .05). CONCLUSION Resistance to nutlin-3 was observed in human bone marrow-derived MSCs; nevertheless, further clinical data are required to be obtained with long-duration exposure to confirm the present findings.
Collapse
Affiliation(s)
- Babak Bajelan
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Majid Zaki-Dizaji
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Rahmani
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sina Darzi
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Shahram Darabi
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Farzad Rajaei
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
25
|
Su QH, Xu XQ, Wang JF, Luan JW, Ren X, Huang HY, Bian SS. Anticancer Effects of Constituents of Herbs Targeting Osteosarcoma. Chin J Integr Med 2019; 25:948-955. [PMID: 31161441 DOI: 10.1007/s11655-019-2941-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2018] [Indexed: 01/04/2023]
Abstract
Osteosarcoma is a rare primary malignancy of bone that is prone to early metastasis. Resection surgery and chemotherapeutic regimens are current standard treatments for osteosarcoma. However, the long-term survival rate of patients with osteosarcoma is low due to a high risk of metastasis. Hence, a new approach is urgently needed to improve the treatment of osteosarcoma. Compared with chemotherapy, natural active constituents isolated from herbs exhibit less adverse effects and better anti-tumor effects. This study aimed to summarize the anticancer effects of constituents of herbs on the progression and metastasis of osteosarcoma cells. It showed that many constituents of herbs inhibited osteosarcoma by targeting proliferation, matrix metalloproteinases, integrin and cadherin, and angiogenesis. The findings might be beneficial for the development of new drugs and treatment strategies.
Collapse
Affiliation(s)
- Qing-Hong Su
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, 250062, China
| | - Xiao-Qun Xu
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, 250062, China
| | - Jun-Fu Wang
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, 250062, China
| | - Jun-Wen Luan
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, 250062, China
| | - Xia Ren
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, 250062, China
| | - Hai-Yan Huang
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, 250062, China
| | - Si-Shan Bian
- Department of Orthopaedics, the Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| |
Collapse
|
26
|
Valente JFA, Sousa A, Queiroz JA, Sousa F. DoE to improve supercoiled p53-pDNA purification by O-phospho-l-tyrosine chromatography. J Chromatogr B Analyt Technol Biomed Life Sci 2018; 1105:184-192. [PMID: 30597418 DOI: 10.1016/j.jchromb.2018.12.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 11/05/2018] [Accepted: 12/02/2018] [Indexed: 02/08/2023]
Abstract
P53 is implicated in various cellular functions and several studies have shown that transfection of cancer cells with wild-type p53-expressing plasmids could directly drive cells into growth arrest and/or apoptosis. In the present work, the 6.07 kbp pcDNA3-FLAG-p53 plasmid, which encodes the p53 tumor suppressor, was produced and recovered from a recombinant cell culture of Escherichia coli DH5α. Following plasmid biosynthesis, the O-phospho-l-tyrosine chromatographic matrix was explored to purify the supercoiled p53-encoding plasmid. In order to quickly determine the optimal chromatographic performance and to obtain the required purity degree, maximizing the recovery yield of the supercoiled plasmid DNA, the Composite Central Face design was applied. The model revealed to be statistically significant (p-value < 0.05), with coefficient of determination of 0.9434 for the recovery yield and 0.9581 for purity and the central point was successfully validated. After the chromatographic process optimization by using the design of experiments tool, 49.7% of the supercoiled p53-encoding plasmid was recovered with 98.2% of purity, when a decreasing ammonium sulphate gradient was applied. The dynamic binding capacity of the O-phospho-l-tyrosine agarose column was 0.35 ± 0.02 mg pDNA/mL matrix at 50% of the breakthrough. Finally, the purified sample was analysed to assess the content of endotoxins, proteins and genomic DNA, showing that all these impurity levels were below the recommendations of the regulatory agencies.
Collapse
Affiliation(s)
- J F A Valente
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - A Sousa
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - J A Queiroz
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - F Sousa
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal.
| |
Collapse
|
27
|
Guo J, Tang Q, Wang Q, Sun W, Pu Z, Wang J, Bao Y. Pifithrin-α enhancing anticancer effect of topotecan on p53-expressing cancer cells. Eur J Pharm Sci 2018; 128:61-72. [PMID: 30472223 DOI: 10.1016/j.ejps.2018.11.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 10/28/2018] [Accepted: 11/21/2018] [Indexed: 01/01/2023]
Abstract
p53 is generally known as an effective anti-cancer molecular, but it is lost or mutated in more than 50% of human tumors. It is still a controversial issue whether the activity of p53 really benefits for treating cancers, we wondered what would happen if the endogenous p53 was inhibited before treated with topotecan (TPT) on p53 positive tumor cells. In this study, pifithrin-α (PFTα), a p53 inhibitor, was used 2 h before treated with TPT on three kinds of cancer cell lines including MCF7, BGC823 and HepG2 cells. The IC50s of TPT for MCF7, BGC823 and HepG2 cells after 10 μΜ PFTα pretreated, was 4.8 to 14.4 folds lower than the effect of TPT alone. It was demonstrated that PFTα decreases the p-p53 levels and p-p53 activity, not affects p53 expression in p53 positive tumor cells. PFTα enhanced anticancer effect of TPT on cells was found mainly by two ways. Firstly, it increased the TPT accumulation in cells and nucleus and promoted the inhibition of TPT on activity of Topo I, and induced more DNA damage. Secondly, PFTα decreased formation of p53/mdm2 complex responsible for p53 degradation by inhibiting the protein expression of mdm2, so p53 degradation was decreased in cytoplasm and p53 accumulation was increased in nucleus, which induced more cells undergo apoptosis. So, the crosstalk between p53 and TPT played a pivotal role for enhancing anticancer effects of PFTα and TPT on p53 positive cancer cells. These findings provide a new idea for drug design and combination chemotherapy of cancers.
Collapse
Affiliation(s)
- Jianli Guo
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, China
| | - Qin Tang
- National Center for Pharmaceutical Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Qingling Wang
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, China
| | - Wenhui Sun
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, China
| | - Zhongji Pu
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, China
| | - Jingyun Wang
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, China
| | - Yongming Bao
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, China; School of Food and Environment Science and Technology, Dalian University of Technology, Panjin 124221, China.
| |
Collapse
|
28
|
Minzel W, Venkatachalam A, Fink A, Hung E, Brachya G, Burstain I, Shaham M, Rivlin A, Omer I, Zinger A, Elias S, Winter E, Erdman PE, Sullivan RW, Fung L, Mercurio F, Li D, Vacca J, Kaushansky N, Shlush L, Oren M, Levine R, Pikarsky E, Snir-Alkalay I, Ben-Neriah Y. Small Molecules Co-targeting CKIα and the Transcriptional Kinases CDK7/9 Control AML in Preclinical Models. Cell 2018; 175:171-185.e25. [PMID: 30146162 DOI: 10.1016/j.cell.2018.07.045] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 07/06/2018] [Accepted: 07/27/2018] [Indexed: 12/22/2022]
Abstract
CKIα ablation induces p53 activation, and CKIα degradation underlies the therapeutic effect of lenalidomide in a pre-leukemia syndrome. Here we describe the development of CKIα inhibitors, which co-target the transcriptional kinases CDK7 and CDK9, thereby augmenting CKIα-induced p53 activation and its anti-leukemic activity. Oncogene-driving super-enhancers (SEs) are highly sensitive to CDK7/9 inhibition. We identified multiple newly gained SEs in primary mouse acute myeloid leukemia (AML) cells and demonstrate that the inhibitors abolish many SEs and preferentially suppress the transcription elongation of SE-driven oncogenes. We show that blocking CKIα together with CDK7 and/or CDK9 synergistically stabilize p53, deprive leukemia cells of survival and proliferation-maintaining SE-driven oncogenes, and induce apoptosis. Leukemia progenitors are selectively eliminated by the inhibitors, explaining their therapeutic efficacy with preserved hematopoiesis and leukemia cure potential; they eradicate leukemia in MLL-AF9 and Tet2-/-;Flt3ITD AML mouse models and in several patient-derived AML xenograft models, supporting their potential efficacy in curing human leukemia.
Collapse
Affiliation(s)
- Waleed Minzel
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Avanthika Venkatachalam
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Avner Fink
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Eric Hung
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Guy Brachya
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Ido Burstain
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Maya Shaham
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Amitai Rivlin
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Itay Omer
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Adar Zinger
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Shlomo Elias
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel; Department of Hematology, Hadassah Medical Center, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Eitan Winter
- Bioinformatics Unit of the I-CORE Computation Center, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | | | | | | | | | | | | | - Nathali Kaushansky
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Liran Shlush
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Moshe Oren
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel
| | - Ross Levine
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eli Pikarsky
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel; Department of Pathology, Hadassah Medical Center, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Irit Snir-Alkalay
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Yinon Ben-Neriah
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel.
| |
Collapse
|
29
|
Maeda A, Nishida Y, Weetall M, Cao L, Branstrom A, Ishizawa J, Nii T, Schober WD, Abe Y, Matsue K, Yoshimura M, Kimura S, Kojima K. Targeting of BMI-1 expression by the novel small molecule PTC596 in mantle cell lymphoma. Oncotarget 2018; 9:28547-28560. [PMID: 29983879 PMCID: PMC6033370 DOI: 10.18632/oncotarget.25558] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 05/21/2018] [Indexed: 12/24/2022] Open
Abstract
Despite the development of the novel Bruton tyrosine kinase inhibitor ibrutinib, mantle cell lymphoma (MCL) remains an incurable B-cell non-Hodgkin lymphoma. BMI-1 is required for the self-renewal and maintenance of MCL-initiating stem cells. Upregulation of BMI-1 has been reported in MCL patients, especially in those with refractory/relapsed disease. We studied the effects of a novel small-molecule selective inhibitor of BMI1 expression, PTC596, in MCL cells. Eight MCL cell lines and patient-derived samples were exposed to PTC596. PTC596 induced mitochondrial apoptosis, as evidenced by loss of mitochondrial membrane potential, caspase-3 cleavage, BAX activation, and phosphatidylserine externalization. There was a positive correlation between baseline BMI-1 protein levels and PTC596-induced apoptosis. p53 status did not affect sensitivity to PTC596. PTC596 effectively decreased BMI-1-expressing and tumor-initiating side population MCL cells (IC50: 138 nM) compared with ibrutinib, which modestly decreased side population cells. Interestingly, PTC596, reported to target cancer stem cells, decreased MCL-1 expression levels and antagonized ibrutinib-induced increase in MCL-1 expression, leading to synergistic apoptosis induction in MCL cells. There are currently no drugs that specifically target cancer stem cell fractions, and a reduction in BMI-1 protein by PTC596 may offer a novel therapeutic strategy for MCL.
Collapse
Affiliation(s)
- Aya Maeda
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Saga University, Saga, Japan
| | - Yuki Nishida
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Saga University, Saga, Japan
| | | | | | | | - Jo Ishizawa
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Takenobu Nii
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wendy D Schober
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yoshiaki Abe
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Kamogawa, Japan
| | - Kosei Matsue
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Kamogawa, Japan
| | - Mariko Yoshimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Saga University, Saga, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Saga University, Saga, Japan
| | - Kensuke Kojima
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Saga University, Saga, Japan
| |
Collapse
|
30
|
Sawyer TK, Partridge AW, Kaan HYK, Juang YC, Lim S, Johannes C, Yuen TY, Verma C, Kannan S, Aronica P, Tan YS, Sherborne B, Ha S, Hochman J, Chen S, Surdi L, Peier A, Sauvagnat B, Dandliker PJ, Brown CJ, Ng S, Ferrer F, Lane DP. Macrocyclic α helical peptide therapeutic modality: A perspective of learnings and challenges. Bioorg Med Chem 2018; 26:2807-2815. [PMID: 29598901 DOI: 10.1016/j.bmc.2018.03.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 03/03/2018] [Accepted: 03/06/2018] [Indexed: 12/20/2022]
Abstract
Macrocyclic α-helical peptides have emerged as a compelling new therapeutic modality to tackle targets confined to the intracellular compartment. Within the scope of hydrocarbon-stapling there has been significant progress to date, including the first stapled α-helical peptide to enter into clinical trials. The principal design concept of stapled α-helical peptides is to mimic a cognate (protein) ligand relative to binding its target via an α-helical interface. However, it was the proclivity of such stapled α-helical peptides to exhibit cell permeability and proteolytic stability that underscored their promise as unique macrocyclic peptide drugs for intracellular targets. This perspective highlights key learnings as well as challenges in basic research with respect to structure-based design, innovative chemistry, cell permeability and proteolytic stability that are essential to fulfill the promise of stapled α-helical peptide drug development.
Collapse
|
31
|
Voltan R, Rimondi E, Melloni E, Rigolin GM, Casciano F, Arcidiacono MV, Celeghini C, Cuneo A, Zauli G, Secchiero P. Ibrutinib synergizes with MDM-2 inhibitors in promoting cytotoxicity in B chronic lymphocytic leukemia. Oncotarget 2018; 7:70623-70638. [PMID: 27661115 PMCID: PMC5342579 DOI: 10.18632/oncotarget.12139] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 09/09/2016] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE The aim of this study was to investigate the anti-leukemic activity of the Bruton tyrosine kinase inhibitor Ibrutinib in combination with the small molecule MDM-2 inhibitor Nutlin-3 in preclinical models. METHODS The potential efficacy of the Ibrutinib/Nutlin-3 combination was evaluated in vitro in a panel of B leukemic cell lines (EHEB, JVM-2, JVM-3, MEC-1, MEC-2) and in primary B-chronic lymphocytic leukemia (B-CLL) patient samples, by assessing cell viability, cell cycle profile, apoptosis and intracellular pathway modulations. Validation of the combination therapy was assessed in a B leukemic xenograft mouse model. RESULTS Ibrutinib exhibited variable anti-leukemic activity in vitro and the combination with Nutlin-3 synergistically enhanced the induction of apoptosis independently from the p53 status. Indeed, the Ibrutinib/Nutlin-3 combination was effective in promoting cytotoxicity also in primary B-CLL samples carrying 17p13 deletion and/or TP53 mutations, already in therapy with Ibrutinib. Molecular analyses performed on both B-leukemic cell lines as well as on primary B-CLL samples, while confirming the switch-off of the MAPK and PI3K pro-survival pathways by Ibrutinib, indicated that the synergism of action with Nutlin-3 was independent by p53 pathway and was accompanied by the activation of the DNA damage cascade signaling through the phosphorylation of the histone protein H2A.X. This observation was confirmed also in the JVM-2 B leukemic xenograft mouse model. CONCLUSIONS Taken together, our data emphasize that the Ibrutinib/Nutlin-3 combination merits to be further evaluated as a therapeutic option for B-CLL.
Collapse
Affiliation(s)
- Rebecca Voltan
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Erika Rimondi
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Elisabetta Melloni
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Gian Matteo Rigolin
- Department of Medical Sciences, Section of Hematology, University of Ferrara, Ferrara, Italy
| | - Fabio Casciano
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Maria Vittoria Arcidiacono
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | | | - Antonio Cuneo
- Department of Medical Sciences, Section of Hematology, University of Ferrara, Ferrara, Italy
| | - Giorgio Zauli
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Paola Secchiero
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| |
Collapse
|
32
|
Prokocimer M, Molchadsky A, Rotter V. Dysfunctional diversity of p53 proteins in adult acute myeloid leukemia: projections on diagnostic workup and therapy. Blood 2017; 130:699-712. [PMID: 28607134 PMCID: PMC5659817 DOI: 10.1182/blood-2017-02-763086] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 06/06/2017] [Indexed: 12/13/2022] Open
Abstract
The heterogeneous nature of acute myeloid leukemia (AML) and its poor prognosis necessitate therapeutic improvement. Current advances in AML research yield important insights regarding AML genetic, epigenetic, evolutional, and clinical diversity, all in which dysfunctional p53 plays a key role. As p53 is central to hematopoietic stem cell functions, its aberrations affect AML evolution, biology, and therapy response and usually predict poor prognosis. While in human solid tumors TP53 is mutated in more than half of cases, TP53 mutations occur in less than one tenth of de novo AML cases. Nevertheless, wild-type (wt) p53 dysfunction due to nonmutational p53 abnormalities appears to be rather frequent in various AML entities, bearing, presumably, a greater impact than is currently appreciated. Hereby, we advocate assessment of adult AML with respect to coexisting p53 alterations. Accordingly, we focus not only on the effects of mutant p53 oncogenic gain of function but also on the mechanisms underlying nonmutational wtp53 inactivation, which might be of therapeutic relevance. Patient-specific TP53 genotyping with functional evaluation of p53 protein may contribute significantly to the precise assessment of p53 status in AML, thus leading to the tailoring of a rationalized and precision p53-based therapy. The resolution of the mechanisms underlying p53 dysfunction will better address the p53-targeted therapies that are currently considered for AML. Additionally, a suggested novel algorithm for p53-based diagnostic workup in AML is presented, aiming at facilitating the p53-based therapeutic choices.
Collapse
MESH Headings
- Adult
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- DNA Damage/drug effects
- Gene Expression Regulation, Leukemic/drug effects
- Genomic Instability/drug effects
- Hematopoiesis/drug effects
- Humans
- Karyopherins/genetics
- Karyopherins/metabolism
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Molecular Targeted Therapy/methods
- Mutation/drug effects
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Nucleophosmin
- Protein Interaction Maps/drug effects
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Signal Transduction/drug effects
- Translocation, Genetic
- Tumor Suppressor Protein p53/analysis
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/metabolism
- Exportin 1 Protein
Collapse
Affiliation(s)
- Miron Prokocimer
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; and
| | - Alina Molchadsky
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel
| | - Varda Rotter
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
33
|
Abstract
Acute lymphoblastic leukemia (ALL) is characterized by a great biological and clinical heterogeneity. Despite most adult patients enter complete hematologic remission after induction therapy only 40% survive five or more years. Over the last 20 years, the definition of an accurate biologic leukemia profile and the minimal residual disease evaluation in addition to conventional risk criteria led to a significant improvement for the risk stratification. The alterations of the oncosuppressor gene TP53, including deletions, sequence mutations and defect in its expression due to regulatory defects, define a new important predictor of adverse outcome. More recently, new drugs have been developed with the aim of targeting p53 protein itself or its regulatory molecules, such as Mdm2, and restoring the pathway functionality. Therefore, TP53 alterations should be considered in the diagnostic work-up to identify high risk ALL patients in need of intensive treatment strategies or eligible for new innovative targeted therapies.
Collapse
Affiliation(s)
- Silvia Salmoiraghi
- a Hematology and Bone Marrow Transplant Unit of Azienda Ospedaliera Papa Giovanni XXIII , Bergamo , Italy
| | - Alessandro Rambaldi
- a Hematology and Bone Marrow Transplant Unit of Azienda Ospedaliera Papa Giovanni XXIII , Bergamo , Italy.,b Department of Hematology-Oncology , University of Milan , Milan , Italy
| | - Orietta Spinelli
- a Hematology and Bone Marrow Transplant Unit of Azienda Ospedaliera Papa Giovanni XXIII , Bergamo , Italy
| |
Collapse
|
34
|
Protein Degradation Systems as Antimalarial Therapeutic Targets. Trends Parasitol 2017; 33:731-743. [PMID: 28688800 DOI: 10.1016/j.pt.2017.05.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 05/23/2017] [Accepted: 05/24/2017] [Indexed: 12/11/2022]
Abstract
Artemisinin (ART)-based combination therapies are the most efficacious treatment of uncomplicated Plasmodium falciparum malaria. Alarmingly, P. falciparum strains have acquired resistance to ART across much of Southeast Asia. ART creates widespread protein and lipid damage inside intraerythrocytic parasites, necessitating macromolecule degradation. The proteasome is the main engine of Plasmodium protein degradation. Indeed, proteasome inhibition and ART have shown synergy in ART-resistant parasites. Moreover, ubiquitin modification is associated with altered parasite susceptibility to multiple antimalarials. Targeting the ubiquitin-proteasome system (UPS), therefore, is an attractive avenue to combat drug resistance. Here, we review recent advances leading to specific targeting of the Plasmodium proteasome. We also highlight the potential for targeting other nonproteasomal protein degradation systems as an additional strategy to disrupt protein homeostasis.
Collapse
|
35
|
Hewings DS, Flygare JA, Bogyo M, Wertz IE. Activity-based probes for the ubiquitin conjugation-deconjugation machinery: new chemistries, new tools, and new insights. FEBS J 2017; 284:1555-1576. [PMID: 28196299 PMCID: PMC7163952 DOI: 10.1111/febs.14039] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/21/2017] [Accepted: 02/10/2017] [Indexed: 12/17/2022]
Abstract
The reversible post‐translational modification of proteins by ubiquitin and ubiquitin‐like proteins regulates almost all cellular processes, by affecting protein degradation, localization, and complex formation. Deubiquitinases (DUBs) are proteases that remove ubiquitin modifications or cleave ubiquitin chains. Most DUBs are cysteine proteases, which makes them well suited for study by activity‐based probes. These DUB probes report on deubiquitinase activity by reacting covalently with the active site in an enzyme‐catalyzed manner. They have proven to be important tools to study DUB selectivity and proteolytic activity in different settings, to identify novel DUBs, and to characterize deubiquitinase inhibitors. Inspired by the efficacy of activity‐based probes for DUBs, several groups have recently reported probes for the ubiquitin conjugation machinery (E1, E2, and E3 enzymes). Many of these enzymes, while not proteases, also posses active site cysteine residues and can be targeted by covalent probes. In this review, we will discuss how features of the probe (cysteine‐reactive group, recognition element, and reporter tag) affect reactivity and suitability for certain experimental applications. We will also review the diverse applications of the current probes, and discuss the need for new probe types to study emerging aspects of ubiquitin biology.
Collapse
Affiliation(s)
- David S Hewings
- Discovery Chemistry, Genentech, South San Francisco, CA, USA.,Early Discovery Biochemistry, Genentech, South San Francisco, CA, USA.,Discovery Oncology, Genentech, South San Francisco, CA, USA.,Department of Pathology, Stanford University School of Medicine, CA, USA
| | - John A Flygare
- Discovery Chemistry, Genentech, South San Francisco, CA, USA
| | - Matthew Bogyo
- Department of Pathology, Stanford University School of Medicine, CA, USA
| | - Ingrid E Wertz
- Early Discovery Biochemistry, Genentech, South San Francisco, CA, USA.,Discovery Oncology, Genentech, South San Francisco, CA, USA
| |
Collapse
|
36
|
Nishida Y, Maeda A, Kim MJ, Cao L, Kubota Y, Ishizawa J, AlRawi A, Kato Y, Iwama A, Fujisawa M, Matsue K, Weetall M, Dumble M, Andreeff M, Davis TW, Branstrom A, Kimura S, Kojima K. The novel BMI-1 inhibitor PTC596 downregulates MCL-1 and induces p53-independent mitochondrial apoptosis in acute myeloid leukemia progenitor cells. Blood Cancer J 2017; 7:e527. [PMID: 28211885 PMCID: PMC5386342 DOI: 10.1038/bcj.2017.8] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 12/20/2016] [Indexed: 12/24/2022] Open
Abstract
Disease recurrence is the major problem in the treatment of acute myeloid leukemia (AML). Relapse is driven by leukemia stem cells, a chemoresistant subpopulation capable of re-establishing disease. Patients with p53 mutant AML are at an extremely high risk of relapse. B-cell-specific Moloney murine leukemia virus integration site 1 (BMI-1) is required for the self-renewal and maintenance of AML stem cells. Here we studied the effects of a novel small molecule inhibitor of BMI-1, PTC596, in AML cells. Treatment with PTC596 reduced MCL-1 expression and triggered several molecular events consistent with induction of mitochondrial apoptosis: loss of mitochondrial membrane potential, BAX conformational change, caspase-3 cleavage and phosphatidylserine externalization. PTC596 induced apoptosis in a p53-independent manner. PTC596 induced apoptosis along with the reduction of MCL-1 and phosphorylated AKT in patient-derived CD34+CD38low/− stem/progenitor cells. Mouse xenograft models demonstrated in vivo anti-leukemia activity of PTC596, which inhibited leukemia cell growth in vivo while sparing normal hematopoietic cells. Our results indicate that PTC596 deserves further evaluation in clinical trials for refractory or relapsed AML patients, especially for those with unfavorable complex karyotype or therapy-related AML that are frequently associated with p53 mutations.
Collapse
Affiliation(s)
- Y Nishida
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Saga University, Saga, Japan
| | - A Maeda
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Saga University, Saga, Japan
| | - M J Kim
- PTC Therapeutics, South Plainfield, NJ, USA
| | - L Cao
- PTC Therapeutics, South Plainfield, NJ, USA
| | - Y Kubota
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Saga University, Saga, Japan
| | - J Ishizawa
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - A AlRawi
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Y Kato
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - A Iwama
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - M Fujisawa
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Kamogawa, Japan
| | - K Matsue
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Kamogawa, Japan
| | - M Weetall
- PTC Therapeutics, South Plainfield, NJ, USA
| | - M Dumble
- Bristol-Myers Squibb, Princeton, NJ, USA
| | - M Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - T W Davis
- PMV Pharmaceuticals Inc., Cranbury, NJ, USA
| | | | - S Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Saga University, Saga, Japan
| | - K Kojima
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Saga University, Saga, Japan
| |
Collapse
|
37
|
Luo ZY, Jiang H, Xu L, Zhang XH. [Rita induce acute lymphoblostic leukemia cell apoptosis by activating P53 pathway]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2017; 38:160-163. [PMID: 28279043 PMCID: PMC7354173 DOI: 10.3760/cma.j.issn.0253-2727.2017.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
| | - H Jiang
- Department of Hematology, Guangzhou Women and Children Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | | | | |
Collapse
|
38
|
Trino S, De Luca L, Laurenzana I, Caivano A, Del Vecchio L, Martinelli G, Musto P. P53-MDM2 Pathway: Evidences for A New Targeted Therapeutic Approach in B-Acute Lymphoblastic Leukemia. Front Pharmacol 2016; 7:491. [PMID: 28018226 PMCID: PMC5159974 DOI: 10.3389/fphar.2016.00491] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/30/2016] [Indexed: 11/13/2022] Open
Abstract
The tumor suppressor p53 is a canonical regulator of different biological functions, like apoptosis, cell cycle arrest, DNA repair, and genomic stability. This gene is frequently altered in human tumors generally by point mutations or deletions. Conversely, in acute lymphoblastic leukemia (ALL) genomic alterations of TP53 are rather uncommon, and prevalently occur in patients at relapse or with poor prognosis. On the other hand, p53 pathway is often compromised by the inactivation of its regulatory proteins, as MDM2 and ARF. MDM2 inhibitor molecules are able to antagonize p53-MDM2 interaction allowing p53 to exert tumor suppressor transcriptional regulation and to induce apoptotic pathways. Recent preclinical and clinical studies propose that MDM2 targeted therapy represents a promising anticancer strategy restoring p53 dependent mechanisms in ALL disease. Here, we discussed the use of new small molecule targeting p53 pathways as a promising drug target therapy in ALL.
Collapse
Affiliation(s)
- Stefania Trino
- Laboratory of Pre-Clinical and Translational Research, IRCCS - Referral Cancer Center of Basilicata Rionero in Vulture (PZ), Italy
| | - Luciana De Luca
- Laboratory of Pre-Clinical and Translational Research, IRCCS - Referral Cancer Center of Basilicata Rionero in Vulture (PZ), Italy
| | - Ilaria Laurenzana
- Laboratory of Pre-Clinical and Translational Research, IRCCS - Referral Cancer Center of Basilicata Rionero in Vulture (PZ), Italy
| | - Antonella Caivano
- Laboratory of Pre-Clinical and Translational Research, IRCCS - Referral Cancer Center of Basilicata Rionero in Vulture (PZ), Italy
| | - Luigi Del Vecchio
- CEINGE - Biotecnologie Avanzate S.C.a R.L.Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, Universita' degli Studi di Napoli Federico IINaples, Italy
| | - Giovanni Martinelli
- Department of Experimental, Diagnostic and Specialty Medicine, Institute of Hematology "L. and A. Seràgnoli," University of Bologna Bologna, Italy
| | - Pellegrino Musto
- Scientific Direction, IRCCS - Referral Cancer Center of Basilicata Rionero in Vulture (PZ), Italy
| |
Collapse
|
39
|
Molecular Changes During Acute Myeloid Leukemia (AML) Evolution and Identification of Novel Treatment Strategies Through Molecular Stratification. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 144:383-436. [PMID: 27865463 DOI: 10.1016/bs.pmbts.2016.09.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Acute myeloid leukemia (AML) is a hematopoietic malignancy characterized by impaired differentiation and uncontrollable proliferation of myeloid progenitor cells. Due to high relapse rates, overall survival for this rapidly progressing disease is poor. The significant challenge in AML treatment is disease heterogeneity stemming from variability in maturation state of leukemic cells of origin, genetic aberrations among patients, and existence of multiple disease clones within a single patient. Disease heterogeneity and the lack of biomarkers for drug sensitivity lie at the root of treatment failure as well as selective efficacy of AML chemotherapies and the emergence of drug resistance. Furthermore, standard-of-care treatment is aggressive, presenting significant tolerability concerns to the commonly advanced-age AML patient. In this review, we examine the concept and potential of molecular stratification, particularly with biologically relevant drug responses, in identifying low-toxicity precision therapeutic combinations and clinically relevant biomarkers for AML patient care as a way to overcome these challenges in AML treatment.
Collapse
|