151
|
Lachowiez CA, Atluri H, DiNardo CD. Advancing the standard: venetoclax combined with intensive induction and consolidation therapy for acute myeloid leukemia. Ther Adv Hematol 2022; 13:20406207221093964. [PMID: 35510212 PMCID: PMC9058453 DOI: 10.1177/20406207221093964] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/28/2022] [Indexed: 01/12/2023] Open
Abstract
The B-cell lymphoma 2 (BCL-2) inhibitor venetoclax (VEN) in combination with lower-intensity therapy is an efficacious treatment for acute myeloid leukemia (AML). VEN in combination with the hypomethylating agent azacitidine improved rates of response and measurable residual disease (MRD)-negative remissions in addition to overall survival in the pivotal phase 3 VIALE-A trial compared with azacitidine monotherapy and has since emerged as the current standard of care in older or unfit patients with AML. In younger, fit patients with AML, intensive induction and consolidation chemotherapy (IC) is commonly employed as frontline therapy; however, relapse remains the principal cause of treatment failure in approximately 30-40% of patients. Improved IC regimens that increase MRD-negative response rates, result in durable remissions, and enable transition to curative allogeneic hematopoietic stem cell transplantation in appropriate patients remain an area of active inquiry. Preliminary results from trials investigating the combination of VEN with IC have reported promising findings to date, with composite complete remission and MRD-negative remission rates of approximately 89-94% and 82-93%, respectively, correlating with improved 12-month event-free and overall survival compared to historical outcomes with IC. Herein, we discuss ongoing trials investigating VEN in combination with IC in addition to outcomes within specific molecularly defined subgroups; review the molecular mechanisms of sensitivity and resistance to VEN, and highlight future combinations of VEN with novel targeted therapies for the treatment of AML.
Collapse
Affiliation(s)
- Curtis A. Lachowiez
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Himachandana Atluri
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Courtney D. DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
152
|
Osterlund EJ, Hirmiz N, Pemberton JM, Nougarède A, Liu Q, Leber B, Fang Q, Andrews DW. Efficacy and specificity of inhibitors of BCL-2 family protein interactions assessed by affinity measurements in live cells. SCIENCE ADVANCES 2022; 8:eabm7375. [PMID: 35442739 PMCID: PMC9020777 DOI: 10.1126/sciadv.abm7375] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/01/2022] [Indexed: 06/14/2023]
Abstract
Cytoplasmic and membrane-bound BCL-2 family proteins regulate apoptosis, a form of programmed cell death, via dozens of binary protein interactions confounding measurement of the effects of inhibitors in live cells. In cancer, apoptosis is frequently dysregulated, and cell survival depends on antiapoptotic proteins binding to and inhibiting proapoptotic BH3 proteins. The clinical success of BH3 mimetic inhibitors of antiapoptotic proteins has spawned major efforts by the pharmaceutical industry to develop molecules with different specificities and higher affinities. Here, quantitative fast fluorescence lifetime imaging microscopy enabled comparison of BH3 mimetic drugs in trials and preclinical development by measuring drug effects on binding affinities of interacting protein pairs in live cells. Both selectivity and efficacy were assessed for 15 inhibitors of four antiapoptotic proteins for each of six BH3 protein ligands. While many drugs target the designed interaction, most also have unexpected selectivity and poor efficacy in cells.
Collapse
Affiliation(s)
- Elizabeth J. Osterlund
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 2J7, Canada
| | - Nehad Hirmiz
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
- School of Biomedical Engineering, McMaster University, Hamilton, Ontario L8S 4L7, Canada
| | - James M. Pemberton
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5S 2J7, Canada
| | - Adrien Nougarède
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
| | - Qian Liu
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
| | - Brian Leber
- Department of Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Qiyin Fang
- School of Biomedical Engineering, McMaster University, Hamilton, Ontario L8S 4L7, Canada
- Department of Engineering Physics, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L7, Canada
| | - David W. Andrews
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 2J7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5S 2J7, Canada
| |
Collapse
|
153
|
Sellin M, Mack R, Rhodes MC, Zhang L, Berg S, Joshi K, Liu S, Wei W, S. J. PB, Larsen P, Taylor RE, Zhang J. Molecular mechanisms by which splice modulator GEX1A inhibits leukaemia development and progression. Br J Cancer 2022; 127:223-236. [PMID: 35422078 PMCID: PMC9296642 DOI: 10.1038/s41416-022-01796-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 02/18/2022] [Accepted: 03/17/2022] [Indexed: 11/09/2022] Open
Abstract
INTRODUCTION Splice modulators have been assessed clinically in treating haematologic malignancies exhibiting splice factor mutations and acute myeloid leukaemia. However, the mechanisms by which such modulators repress leukaemia remain to be elucidated. OBJECTIVES The primary goal of this assessment was to assess the molecular mechanism by which the natural splice modulator GEX1A kills leukaemic cells in vitro and within in vivo mouse models. METHODS Using human leukaemic cell lines, we assessed the overall sensitivity these cells have to GEX1A via EC50 analysis. We subsequently analysed its effects using in vivo xenograft mouse models and examined whether cell sensitivities were correlated to genetic characteristics or protein expression levels. We also utilised RT-PCR and RNAseq analyses to determine splice change and RNA expression level differences between sensitive and resistant leukaemic cell lines. RESULTS We found that, in vitro, GEX1A induced an MCL-1 isoform shift to pro-apoptotic MCL-1S in all leukaemic cell types, though sensitivity to GEX1A-induced apoptosis was negatively associated with BCL-xL expression. In BCL-2-expressing leukaemic cells, GEX1A induced BCL-2-dependent apoptosis by converting pro-survival BCL-2 into a cell killer. Thus, GEX1A + selective BCL-xL inhibition induced synergism in killing leukaemic cells, while GEX1A + BCL-2 inhibition showed antagonism in BCL-2-expressing leukaemic cells. In addition, GEX1A sensitised FLT3-ITD+ leukaemic cells to apoptosis by inducing aberrant splicing and repressing the expression of FLT3-ITD. Consistently, in in vivo xenografts, GEX1A killed the bulk of leukaemic cells via apoptosis when combined with BCL-xL inhibition. Furthermore, GEX1A repressed leukaemia development by targeting leukaemia stem cells through inhibiting FASTK mitochondrial isoform expression across sensitive and non-sensitive leukaemia types. CONCLUSION Our study suggests that GEX1A is a potent anti-leukaemic agent in combination with BCL-xL inhibitors, which targets leukaemic blasts and leukaemia stem cells through distinct mechanisms.
Collapse
|
154
|
Huang D, Wang Y, Hu B, Luo Z, Huang J, Wang J, Zhang F. A computational perspective for tailor-made selective Mcl-1 and Bcl-XL inhibitors. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.132269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
155
|
Wu G, Zhang C, Xu L, Chen H, Fan X, Sun B, Tang Q, Zhan Y, Chen T, Wang X. BAK plays a key role in A-1331852-induced apoptosis in senescent chondrocytes. Biochem Biophys Res Commun 2022; 609:93-99. [PMID: 35421634 DOI: 10.1016/j.bbrc.2022.03.155] [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: 03/02/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 11/02/2022]
Abstract
Osteoarthritis occurs when the number of senescent chondrocytes in the joints reaches an intolerable level. The purpose of our study was to explore the therapeutic effect and mechanism of action of A-1331852 in osteoarthritis. Doxorubicin and etoposide were used to induce cell senescence as determined by the cessation of cell proliferation, augmented senescence-associated beta-galactosidase (SA-β-Gal) staining, and increased p53 expression levels. The CCK-8 cytotoxicity assay and SA-β-Gal staining demonstrated that Bcl-xL inhibitors could selectively remove senescent chondrocytes without damaging healthy chondrocytes. A-1331852 induced caspase-dependent death of senescent chondrocytes with decreased mitochondrial membrane potential, nuclear concentration, plasma membrane rupture, and PARP cleavage. Most importantly, A-1331852 upregulated BAK expression levels, indicating that BAK plays a key role in the A-1331852-induced apoptosis of senescent chondrocytes. Live-cell fluorescence resonance energy transfer showed that A-1331852 detached the binding of Bcl-xL to BAK and promoted the oligomerization of BAK on the mitochondrial membrane. In conclusion, this study provides the first evidence that A-1331852 selectively promotes apoptosis in senescent chondrocytes by interfering with the interaction between Bcl-xL and BAK.
Collapse
Affiliation(s)
- Guihao Wu
- Department of Pain Management, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Cifeng Zhang
- Department of Pain Management, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Lingjun Xu
- Department of Pain Management, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Hongce Chen
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Xuhong Fan
- Department of Pain Management, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Beini Sun
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Qiling Tang
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Yongtong Zhan
- Department of Pain Management, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Tongsheng Chen
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Xiaoping Wang
- Department of Pain Management, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.
| |
Collapse
|
156
|
Xu W, Zhao T, Chen H, Huang N, Gong H, Zhang J, Yang Y, Li T, Zhang G, Gong C, Yang M, Xiao H. Pan-mTOR inhibitors sensitize the senolytic activity of Navitoclax via mTORC2 inhibition-mediated apoptotic signaling. Biochem Pharmacol 2022; 200:115045. [DOI: 10.1016/j.bcp.2022.115045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/09/2022] [Accepted: 04/12/2022] [Indexed: 11/30/2022]
|
157
|
Targeting oncogene and non-oncogene addiction to inflame the tumour microenvironment. Nat Rev Drug Discov 2022; 21:440-462. [PMID: 35292771 DOI: 10.1038/s41573-022-00415-5] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2022] [Indexed: 12/12/2022]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the clinical management of multiple tumours. However, only a few patients respond to ICIs, which has generated considerable interest in the identification of resistance mechanisms. One such mechanism reflects the ability of various oncogenic pathways, as well as stress response pathways required for the survival of transformed cells (a situation commonly referred to as 'non-oncogene addiction'), to support tumour progression not only by providing malignant cells with survival and/or proliferation advantages, but also by establishing immunologically 'cold' tumour microenvironments (TMEs). Thus, both oncogene and non-oncogene addiction stand out as promising targets to robustly inflame the TME and potentially enable superior responses to ICIs.
Collapse
|
158
|
Morsli S, Doherty GJ, Muñoz-Espín D. Activatable senoprobes and senolytics: Novel strategies to detect and target senescent cells. Mech Ageing Dev 2022; 202:111618. [PMID: 34990647 DOI: 10.1016/j.mad.2021.111618] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/21/2021] [Accepted: 12/24/2021] [Indexed: 01/10/2023]
Abstract
Pharmacologically active compounds that manipulate cellular senescence (senotherapies) have recently shown great promise in multiple pre-clinical disease models, and some of them are now being tested in clinical trials. Despite promising proof-of-principle evidence, there are known on- and off-target toxicities associated with these compounds, and therefore more refined and novel strategies to improve their efficacy and specificity for senescent cells are being developed. Preferential release of drugs and macromolecular formulations within senescent cells has been predominantly achieved by exploiting one of the most widely used biomarkers of senescence, the increase in lysosomal senescence-associated β-galactosidase (SA-β-gal) activity, a common feature of most reported senescent cell types. Galacto-conjugation is a versatile therapeutic and detection strategy to facilitate preferential targeting of senescent cells by using a variety of existing formulations, including modular systems, nanocarriers, activatable prodrugs, probes, and small molecules. We discuss the benefits and drawbacks of these specific senescence targeting tools and how the strategy of galacto-conjugation might be utilised to design more specific and sophisticated next-generation senotherapeutics, as well as theranostic agents. Finally, we discuss some innovative strategies and possible future directions for the field.
Collapse
Affiliation(s)
- Samir Morsli
- CRUK Cambridge Centre Early Detection Programme, Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, UK
| | - Gary J Doherty
- Department of Oncology, Box 193, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK.
| | - Daniel Muñoz-Espín
- CRUK Cambridge Centre Early Detection Programme, Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, UK.
| |
Collapse
|
159
|
Zehnle PMA, Wu Y, Pommerening H, Erlacher M. Stayin‘ alive: BCL-2 proteins in the hematopoietic system. Exp Hematol 2022; 110:1-12. [PMID: 35315320 DOI: 10.1016/j.exphem.2022.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 11/04/2022]
|
160
|
Scott SC, Anders NM, He P, Hemingway A, Gore SD, Hann CL, Rudek MA. Validation of a robust and rapid liquid chromatography tandem mass spectrometric method for the quantitative analysis of navitoclax. Biomed Chromatogr 2022; 36:e5289. [PMID: 34846750 PMCID: PMC8881338 DOI: 10.1002/bmc.5289] [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: 11/01/2021] [Revised: 11/13/2021] [Accepted: 11/15/2021] [Indexed: 11/05/2022]
Abstract
The Bcl-2 family small molecule inhibitor navitoclax is being clinically evaluated to treat multiple cancers including lymphoid malignancies and small cell lung cancer. A sensitive and reliable method was developed to quantitate navitoclax in human plasma using liquid chromatography with tandem mass spectrometry with which to perform detailed pharmacokinetic studies. Sample preparation involved protein precipitation using acetonitrile. Separation of navitoclax and the internal standard, navitoclax-d8, was achieved with a Waters Acquity UPLC BEH C18 column using isocratic flow over a 3 min total analytical run time. A SCIEX 4500 triple quadrupole mass spectrometer operated in positive electrospray ionization mode was used for the detection of navitoclax. The assay range was 5-5,000 ng/ml and proved to be accurate (89.5-104.9%) and precise (CV ≤ 11%). Long-term frozen plasma stability for navitoclax at -70°C was at least 34 months. The method was applied for the measurement of total plasma concentration of navitoclax in a patient receiving a 250 mg daily oral dose.
Collapse
Affiliation(s)
- Susan C. Scott
- Department of Oncology, School of Medicine, Johns Hopkins University, 1650 Orleans Street, Baltimore, Maryland, 21231 USA,The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Nicole M. Anders
- Department of Oncology, School of Medicine, Johns Hopkins University, 1650 Orleans Street, Baltimore, Maryland, 21231 USA,The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Ping He
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Avelina Hemingway
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Steven D. Gore
- IDB/CTEP/NCI, National Cancer Institute, Rockville, Maryland
| | - Christine L. Hann
- Department of Oncology, School of Medicine, Johns Hopkins University, 1650 Orleans Street, Baltimore, Maryland, 21231 USA,The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA,Correspondence to: Michelle A. Rudek, PharmD, PhD, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine,1650 Orleans Street, CRB1 Room 1M52, Baltimore, MD 21287, Phone: 443-287-6476, and Christine L. Hann, MD, PhD, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Viragh 8123 Box 6, 201 N. Broadway, Baltimore, MD 21287, Phone: 443-287-6476,
| | - Michelle A. Rudek
- Department of Oncology, School of Medicine, Johns Hopkins University, 1650 Orleans Street, Baltimore, Maryland, 21231 USA,The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA,Division of Clinical Pharmacology, Department of Medicine, School of Medicine, Johns Hopkins University, 1650 Orleans Street, Baltimore, Maryland, 21231 USA,Correspondence to: Michelle A. Rudek, PharmD, PhD, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine,1650 Orleans Street, CRB1 Room 1M52, Baltimore, MD 21287, Phone: 443-287-6476, and Christine L. Hann, MD, PhD, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Viragh 8123 Box 6, 201 N. Broadway, Baltimore, MD 21287, Phone: 443-287-6476,
| |
Collapse
|
161
|
Rayson A, Boudiffa M, Naveed M, Griffin J, Dall’Ara E, Bellantuono I. Geroprotectors and Skeletal Health: Beyond the Headlines. Front Cell Dev Biol 2022; 10:682045. [PMID: 35223825 PMCID: PMC8864221 DOI: 10.3389/fcell.2022.682045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 01/10/2022] [Indexed: 12/19/2022] Open
Abstract
Osteoporosis and osteoarthritis are the most common age-related diseases of the musculoskeletal system. They are responsible for high level of healthcare use and are often associated with comorbidities. Mechanisms of ageing such as senescence, inflammation and autophagy are common drivers for both diseases and molecules targeting those mechanisms (geroprotectors) have potential to prevent both diseases and their co-morbidities. However, studies to test the efficacy of geroprotectors on bone and joints are scant. The limited studies available show promising results to prevent and reverse Osteoporosis-like disease. In contrast, the effects on the development of Osteoarthritis-like disease in ageing mice has been disappointing thus far. Here we review the literature and report novel data on the effect of geroprotectors for Osteoporosis and Osteoarthritis, we challenge the notion that extension of lifespan correlates with extension of healthspan in all tissues and we highlight the need for more thorough studies to test the effects of geroprotectors on skeletal health in ageing organisms.
Collapse
Affiliation(s)
- Alexandra Rayson
- Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, Sheffield, United Kingdom
| | - Maya Boudiffa
- Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, Sheffield, United Kingdom
| | - Maneeha Naveed
- Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, Sheffield, United Kingdom
| | - Jon Griffin
- Healthy Lifespan Institute, Department of Molecular Biology and Biotechnology, The University of Sheffield, Sheffield, United Kingdom
| | - Enrico Dall’Ara
- Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, Sheffield, United Kingdom
- Insigneo Institute for in silico Medicine, Sheffield, United Kingdom
| | - Ilaria Bellantuono
- Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, Sheffield, United Kingdom
| |
Collapse
|
162
|
Joly F, Fabbro M, Follana P, Lequesne J, Medioni J, Lesoin A, Frenel JS, Abadie-Lacourtoisie S, Floquet A, Gladieff L, You B, Gavoille C, Kalbacher E, Briand M, Brachet PE, Giffard F, Weiswald LB, Just PA, Blanc-Fournier C, Leconte A, Clarisse B, Leary A, Poulain L. A phase II study of Navitoclax (ABT-263) as single agent in women heavily pretreated for recurrent epithelial ovarian cancer: The MONAVI – GINECO study. Gynecol Oncol 2022; 165:30-39. [DOI: 10.1016/j.ygyno.2022.01.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/14/2022] [Accepted: 01/16/2022] [Indexed: 12/31/2022]
|
163
|
Li L, Li P, Song H, Ma X, Zeng S, Peng Y, Zhang G. Targeting entry into mitochondria for increased anticancer efficacy of BCL-X L-selective inhibitors in lung cancer. Pharmacol Res 2022; 177:106095. [PMID: 35074525 DOI: 10.1016/j.phrs.2022.106095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 11/25/2022]
Abstract
The BCL-XL-selective inhibitors exhibit potential clinical application value when combined with chemotherapeutic drugs for the treatment of solid tumors. However, their efficacy in these settings is still low when treated with BCL-XL inhibitors alone in solid tumors. The mechanism responsible for the poor efficacy remains unclear. We show here that unable to interact with target of BCL-XL-selective inhibitors caused by invalid entry into mitochondria is essential for their inefficacy in solid tumors. We demonstrated in non-small-cell lung cancer (NSCLC) cells that the instability of A-1155463 in cells as well as invalid entry into mitochondria of A-1331852, two BCL-XL-selective inhibitors, accounted for their off-target problems. Furthermore, we found that a mitochondria-targeted, non-toxic small molecule NA-2a improved the on-target effect of A-1331852 to enhance its apoptotic regulatory activity, thereby increasing its anticancer activity in NSCLC. Our results indicated that NA-2a was selectively enriched in mitochondria transported by organic-anion-transporting polypeptide (OATP) transporters, which altered the permeability of the mitochondrial membrane, thereby promoting the entrance of A-1331852 to mitochondria and enhancing its disruption of the BIM-BCL-XL complex, which finally led to the increased anticancer activity in vitro and in vivo. Collectively, our data provided overwhelming evidence that the combination of NA-2a and A-1331852 could be used as a promising synergistic therapeutic agent in NSCLC therapy.
Collapse
Affiliation(s)
- Liangping Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Pingping Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Huanhuan Song
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Xuesong Ma
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Shulan Zeng
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| | - Yan Peng
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| | - Guohai Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| |
Collapse
|
164
|
Westhoff MA, Schuler-Ortoli M, Zerrinius D, Hadzalic A, Schuster A, Strobel H, Scheuerle A, Wong T, Wirtz CR, Debatin KM, Peraud A. Bcl-XL but Not Bcl-2 Is a Potential Target in Medulloblastoma Therapy. Pharmaceuticals (Basel) 2022; 15:ph15010091. [PMID: 35056150 PMCID: PMC8779796 DOI: 10.3390/ph15010091] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/29/2021] [Accepted: 01/05/2022] [Indexed: 01/26/2023] Open
Abstract
Medulloblastoma (MB) is the most common solid tumour in children and, despite current treatment with a rather aggressive combination therapy, accounts for 10% of all deaths associated with paediatric cancer. Breaking the tumour cells’ intrinsic resistance to therapy-induced cell death should lead to less aggressive and more effective treatment options. In other tumour entities, this has been achieved by modulating the balance between the various pro- and anti-apoptotic members of the Bcl-2 family with small molecule inhibitors. To evaluate the therapeutic benefits of ABT-199 (Venetoclax), a Bcl-2 inhibitor, and ABT-263 (Navitoclax), a dual Bcl-XL/Bcl-2 inhibitor, increasingly more relevant model systems were investigated. Starting from established MB cell lines, progressing to primary patient-derived material and finally an experimental tumour system imbedded in an organic environment were chosen. Assessment of the metabolic activity (a surrogate readout for population viability), the induction of DNA fragmentation (apoptosis) and changes in cell number (the combined effect of alterations in proliferation and cell death induction) revealed that ABT-263, but not ABT-199, is a promising candidate for combination therapy, synergizing with cell death-inducing stimuli. Interestingly, in the experimental tumour setting, the sensitizing effect of ABT-263 seems to be predominantly mediated via an anti-proliferative and not a pro-apoptotic effect, opening a future line of investigation. Our data show that modulation of specific members of the Bcl-2 family might be a promising therapeutic addition for the treatment of MB.
Collapse
Affiliation(s)
- Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, Ulm University Hospital, 89075 Ulm, Germany; (A.S.); (H.S.); (T.W.); (K.-M.D.)
- Correspondence: (M.-A.W.); (A.P.); Tel.: +49-731-500-57495 (M.-A.W.); +49-731-500-55001 (A.P.)
| | - Marie Schuler-Ortoli
- Section Pediatric Neurosurgery, Department of Neurosurgery, Ulm University Hospital, 89081 Ulm, Germany; (M.S.-O.); (D.Z.); (A.H.)
| | - Daniela Zerrinius
- Section Pediatric Neurosurgery, Department of Neurosurgery, Ulm University Hospital, 89081 Ulm, Germany; (M.S.-O.); (D.Z.); (A.H.)
| | - Amina Hadzalic
- Section Pediatric Neurosurgery, Department of Neurosurgery, Ulm University Hospital, 89081 Ulm, Germany; (M.S.-O.); (D.Z.); (A.H.)
| | - Andrea Schuster
- Department of Pediatrics and Adolescent Medicine, Ulm University Hospital, 89075 Ulm, Germany; (A.S.); (H.S.); (T.W.); (K.-M.D.)
| | - Hannah Strobel
- Department of Pediatrics and Adolescent Medicine, Ulm University Hospital, 89075 Ulm, Germany; (A.S.); (H.S.); (T.W.); (K.-M.D.)
| | | | - Tiana Wong
- Department of Pediatrics and Adolescent Medicine, Ulm University Hospital, 89075 Ulm, Germany; (A.S.); (H.S.); (T.W.); (K.-M.D.)
- Section Pediatric Neurosurgery, Department of Neurosurgery, Ulm University Hospital, 89081 Ulm, Germany; (M.S.-O.); (D.Z.); (A.H.)
| | | | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, Ulm University Hospital, 89075 Ulm, Germany; (A.S.); (H.S.); (T.W.); (K.-M.D.)
| | - Aurelia Peraud
- Section Pediatric Neurosurgery, Department of Neurosurgery, Ulm University Hospital, 89081 Ulm, Germany; (M.S.-O.); (D.Z.); (A.H.)
- Correspondence: (M.-A.W.); (A.P.); Tel.: +49-731-500-57495 (M.-A.W.); +49-731-500-55001 (A.P.)
| |
Collapse
|
165
|
Diepstraten ST, Anderson MA, Czabotar PE, Lessene G, Strasser A, Kelly GL. The manipulation of apoptosis for cancer therapy using BH3-mimetic drugs. Nat Rev Cancer 2022; 22:45-64. [PMID: 34663943 DOI: 10.1038/s41568-021-00407-4] [Citation(s) in RCA: 203] [Impact Index Per Article: 67.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/02/2021] [Indexed: 12/14/2022]
Abstract
Apoptosis is a form of programmed cell death that is regulated by the balance between prosurvival and proapoptotic BCL-2 protein family members. Evasion of apoptosis is a hallmark of cancer that arises when this balance is tipped in favour of survival. One form of anticancer therapeutic, termed 'BH3-mimetic drugs', has been developed to directly activate the apoptosis machinery in malignant cells. These drugs bind to and inhibit specific prosurvival BCL-2 family proteins, thereby mimicking their interaction with the BH3 domains of proapoptotic BCL-2 family proteins. The BCL-2-specific inhibitor venetoclax is approved by the US Food and Drug Administration and many regulatory authorities worldwide for the treatment of chronic lymphocytic leukaemia and acute myeloid leukaemia. BH3-mimetic drugs targeting other BCL-2 prosurvival proteins have been tested in preclinical models of cancer, and drugs targeting MCL-1 or BCL-XL have advanced into phase I clinical trials for certain cancers. As with all therapeutics, efficacy and tolerability need to be carefully balanced to achieve a therapeutic window whereby there is significant anticancer activity with an acceptable safety profile. In this Review, we outline the current state of BH3-mimetic drugs targeting various prosurvival BCL-2 family proteins and discuss emerging data regarding primary and acquired resistance to these agents and approaches that may overcome this. We highlight issues that need to be addressed to further advance the clinical application of BH3-mimetic drugs, both alone and in combination with additional anticancer agents (for example, standard chemotherapeutic drugs or inhibitors of oncogenic kinases), for improved responses in patients with cancer.
Collapse
Affiliation(s)
- Sarah T Diepstraten
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Mary Ann Anderson
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
- Department of Clinical Haematology, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Peter E Czabotar
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Guillaume Lessene
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
- Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, VIC, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia.
| | - Gemma L Kelly
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
166
|
Lim S, An SB, Jung M, Joshi HP, Kumar H, Kim C, Song SY, Lee J, Kang M, Han I, Kim B. Local Delivery of Senolytic Drug Inhibits Intervertebral Disc Degeneration and Restores Intervertebral Disc Structure. Adv Healthc Mater 2022; 11:e2101483. [PMID: 34699690 DOI: 10.1002/adhm.202101483] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/09/2021] [Indexed: 12/25/2022]
Abstract
Intervertebral disc (IVD) degeneration (IVDD) is a leading cause of chronic low back pain. There is a strong clinical demand for more effective treatments for IVDD as conventional treatments provide only symptomatic relief rather than arresting IVDD progression. This study shows that senolytic therapy with local drug delivery can inhibit IVDD and restore IVD integrity. ABT263, a senolytic drug, is loaded in poly(lactic-co-glycolic acid) nanoparticles (PLGA-ABT) and intradiscally administered into injury-induced IVDD rat models. The single intradiscal injection of PLGA-ABT may enable local delivery of the drug to avascular IVD, prevention of potential systemic toxicity caused by systemic administration of senolytic drug, and morbidity caused by repetitive injections of free drug into the IVD. The strategy results in the selective elimination of senescent cells from the degenerative IVD, reduces expressions of pro-inflammatory cytokines and matrix proteases in the IVD, inhibits progression of IVDD, and even restores the IVD structure. This study demonstrates for the first time that local delivery of senolytic drug can effectively treat senescence-associated IVDD. This approach can be extended to treat other types of senescence-associated degenerative diseases.
Collapse
Affiliation(s)
- Songhyun Lim
- School of Chemical and Biological Engineering Seoul National University Seoul 08826 Republic of Korea
| | - Seong Bae An
- Department of Neurosurgery CHA University School of Medicine CHA Bundang Medical Center, Seongnam‐si Gyeonggi‐do 13496 Republic of Korea
| | - Mungyo Jung
- School of Chemical and Biological Engineering Seoul National University Seoul 08826 Republic of Korea
| | - Hari Prasad Joshi
- Department of Neurosurgery CHA University School of Medicine CHA Bundang Medical Center, Seongnam‐si Gyeonggi‐do 13496 Republic of Korea
| | - Hemant Kumar
- Department of Pharmacology and Toxicology National Institute of Pharmaceutical Education and Research (NIPER)‐Ahmedabad Gandhinagar Gujarat 382355 India
| | - Cheesue Kim
- School of Chemical and Biological Engineering Seoul National University Seoul 08826 Republic of Korea
| | - Seuk Young Song
- School of Chemical and Biological Engineering Seoul National University Seoul 08826 Republic of Korea
| | - Ju‐Ro Lee
- School of Chemical and Biological Engineering Seoul National University Seoul 08826 Republic of Korea
| | - Mikyung Kang
- Interdisciplinary Program for Bioengineering Seoul National University Seoul 08826 Republic of Korea
| | - Inbo Han
- Department of Neurosurgery CHA University School of Medicine CHA Bundang Medical Center, Seongnam‐si Gyeonggi‐do 13496 Republic of Korea
| | - Byung‐Soo Kim
- School of Chemical and Biological Engineering Seoul National University Seoul 08826 Republic of Korea
- Interdisciplinary Program for Bioengineering Seoul National University Seoul 08826 Republic of Korea
- Institute of Chemical Processes Institute of Engineering Research BioMAX Seoul National University Seoul 08826 Republic of Korea
| |
Collapse
|
167
|
Mitochondria-mediated oxidative stress during viral infection. Trends Microbiol 2022; 30:679-692. [DOI: 10.1016/j.tim.2021.12.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/20/2022]
|
168
|
Luan J, Hu B, Wang S, Liu H, Lu S, Li W, Sun X, Shi J, Wang J. Selectivity mechanism of BCL-XL/2 inhibition through in silico investigation. Phys Chem Chem Phys 2022; 24:17105-17115. [DOI: 10.1039/d2cp01755e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
BCL-XL protein is among the most important members of the anti-apoptotic subfamily of BCL-2 protein family, as currently a promising new target for anti-tumor drug research, even though BCL-XL/2 proteins...
Collapse
|
169
|
Westaby D, Jimenez-Vacas JM, Padilha A, Varkaris A, Balk SP, de Bono JS, Sharp A. Targeting the Intrinsic Apoptosis Pathway: A Window of Opportunity for Prostate Cancer. Cancers (Basel) 2021; 14:51. [PMID: 35008216 PMCID: PMC8750516 DOI: 10.3390/cancers14010051] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/12/2021] [Accepted: 12/15/2021] [Indexed: 12/15/2022] Open
Abstract
Despite major improvements in the management of advanced prostate cancer over the last 20 years, the disease remains invariably fatal, and new effective therapies are required. The development of novel hormonal agents and taxane chemotherapy has improved outcomes, although primary and acquired resistance remains problematic. Inducing cancer cell death via apoptosis has long been an attractive goal in the treatment of cancer. Apoptosis, a form of regulated cell death, is a highly controlled process, split into two main pathways (intrinsic and extrinsic), and is stimulated by a multitude of factors, including cellular and genotoxic stress. Numerous therapeutic strategies targeting the intrinsic apoptosis pathway are in clinical development, and BH3 mimetics have shown promising efficacy for hematological malignancies. Utilizing these agents for solid malignancies has proved more challenging, though efforts are ongoing. Molecular characterization and the development of predictive biomarkers is likely to be critical for patient selection, by identifying tumors with a vulnerability in the intrinsic apoptosis pathway. This review provides an up-to-date overview of cell death and apoptosis, specifically focusing on the intrinsic pathway. It summarizes the latest approaches for targeting the intrinsic apoptosis pathway with BH3 mimetics and discusses how these strategies may be leveraged to treat prostate cancer.
Collapse
Affiliation(s)
- Daniel Westaby
- Division of Clinical Studies, The Institute of Cancer Research, London SM2 5NG, UK; (D.W.); (J.M.J.-V.); (A.P.) (J.S.d.B.)
- Prostate Cancer Targeted Therapy Group, The Royal Marsden Hospital, London SM2 5PT, UK
| | - Juan M. Jimenez-Vacas
- Division of Clinical Studies, The Institute of Cancer Research, London SM2 5NG, UK; (D.W.); (J.M.J.-V.); (A.P.) (J.S.d.B.)
| | - Ana Padilha
- Division of Clinical Studies, The Institute of Cancer Research, London SM2 5NG, UK; (D.W.); (J.M.J.-V.); (A.P.) (J.S.d.B.)
| | - Andreas Varkaris
- Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (A.V.); (S.P.B.)
| | - Steven P. Balk
- Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (A.V.); (S.P.B.)
| | - Johann S. de Bono
- Division of Clinical Studies, The Institute of Cancer Research, London SM2 5NG, UK; (D.W.); (J.M.J.-V.); (A.P.) (J.S.d.B.)
- Prostate Cancer Targeted Therapy Group, The Royal Marsden Hospital, London SM2 5PT, UK
| | - Adam Sharp
- Division of Clinical Studies, The Institute of Cancer Research, London SM2 5NG, UK; (D.W.); (J.M.J.-V.); (A.P.) (J.S.d.B.)
- Prostate Cancer Targeted Therapy Group, The Royal Marsden Hospital, London SM2 5PT, UK
| |
Collapse
|
170
|
Abstract
DNA mutation is a common event in the human body, but in most situations, it is fixed right away by the DNA damage response program. In case the damage is too severe to repair, the programmed cell death system will be activated to get rid of the cell. However, if the damage affects some critical components of this system, the genetic scars are kept and multiply through mitosis, possibly leading to cancer someday. There are many forms of programmed cell death, but apoptosis and necroptosis represent the default and backup strategy, respectively, in the maintenance of optimal cell population as well as in cancer prevention. For the same reason, the ideal approach for cancer treatment is to induce apoptosis in the cancer cells because it proceeds 20 times faster than tumor cell proliferation and leaves no mess behind. Induction of necroptosis can be the second choice in case apoptosis becomes hard to achieve, however, necroptosis finishes the job at a cost-inflammation.
Collapse
Affiliation(s)
- Xianmei Meng
- Inner Mongolia Institute of Digestive Diseases, Inner Mongolia Engineering Research Center for Prevention and Treatment of Digestive Diseases, Inner Mongolia University of Science and Technology, 74506The Second Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Tong Dang
- Inner Mongolia Institute of Digestive Diseases, Inner Mongolia Engineering Research Center for Prevention and Treatment of Digestive Diseases, Inner Mongolia University of Science and Technology, 74506The Second Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Jianyuan Chai
- Inner Mongolia Institute of Digestive Diseases, Inner Mongolia Engineering Research Center for Prevention and Treatment of Digestive Diseases, Inner Mongolia University of Science and Technology, 74506The Second Affiliated Hospital of Baotou Medical College, Baotou, China.,Laboratory of Gastrointestinal Injury and Cancer, VA Long Beach Healthcare System, Long Beach, CA, USA.,College of Medicine, University of California, Irvine, CA, USA
| |
Collapse
|
171
|
Senescent tumor cells: an overlooked adversary in the battle against cancer. Exp Mol Med 2021; 53:1834-1841. [PMID: 34916607 PMCID: PMC8741813 DOI: 10.1038/s12276-021-00717-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/07/2021] [Accepted: 09/14/2021] [Indexed: 01/10/2023] Open
Abstract
AbstractSenescent cells in cancer tissue, including senescent fibroblasts and macrophages, have been reported to increase the malignant potency of cancer cells by secreting senescence-associated secretory phenotype (SASP). Otherwise, Senescence of tumor cells has been believed to inhibit tumor growth by halting the massive proliferation and increasing the chances of immune clearance. In particular, senescent tumor cells (STCs) have been thought that they rarely exist in carcinomas because oncogene-induced senescence needs to be overcome for protumorigenic cells to become malignant. However, recent studies have revealed that a considerable number of STCs are present in cancer tissue, even in metastatic sites. In fact, STCs are widely involved in cancer progression by leading to collective invasion and building a cytokine barrier to protect nonsenescent tumor cells from immune attack. Furthermore, therapy-induced STCs can induce tumor progression and recurrence by increasing stemness. However, obscure causative factors and their heterogeneity in various cancers make it difficult to establish the physiological role of STCs. Here, we summarize and review the current knowledge of the pathophysiology and role of STCs. We also outline the current status of therapeutic strategies for directly removing STCs or modulating the SASPs to maximize the positive functions of STCs while suppressing the negative functions.
Collapse
|
172
|
Bleijs M, Pleijte C, Engels S, Ringnalda F, Meyer-Wentrup F, van de Wetering M, Clevers H. EWSR1-WT1 Target Genes and Therapeutic Options Identified in a Novel DSRCT In Vitro Model. Cancers (Basel) 2021; 13:cancers13236072. [PMID: 34885181 PMCID: PMC8657306 DOI: 10.3390/cancers13236072] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 11/24/2021] [Accepted: 11/28/2021] [Indexed: 11/28/2022] Open
Abstract
Simple Summary Desmoplastic small round cell tumor (DSRCT) is an extremely rare soft tissue sarcoma arising in the abdomen of adolescents and young adults. This sarcoma is driven by a single genomic rearrangement, resulting in the expression of the EWSR1-WT1 fusion gene. No effective treatment exists for DSRCT patients, which highlights the need for preclinical models to study disease progression and drug sensitivity. The aim of this study is to develop a pre-clinical DSRCT in vitro model, which enables investigating the molecular target genes of the EWSR1-WT1 fusion gene and allows for medium-throughput drug screening to discover new therapeutic options. Abstract Desmoplastic small round cell tumor (DSRCT) is a rare and aggressive soft tissue sarcoma with a lack of effective treatment options and a poor prognosis. DSRCT is characterized by a chromosomal translocation, resulting in the EWSR1-WT1 gene fusion. The molecular mechanisms driving DSRCT are poorly understood, and a paucity of preclinical models hampers DSRCT research. Here, we establish a novel primary patient-derived DSRCT in vitro model, recapitulating the original tumor. We find that EWSR1-WT1 expression affects cell shape and cell survival, and we identify downstream target genes of the EWSR1-WT1 fusion. Additionally, this preclinical in vitro model allows for medium-throughput drug screening. We discover sensitivity to several drugs, including compounds targeting RTKs. MERTK, which has been described as a therapeutic target for several malignancies, correlates with EWSR1-WT1 expression. Inhibition of MERTK with the small-molecule inhibitor UNC2025 results in reduced proliferation of DSRCT cells in vitro, suggesting MERTK as a therapeutic target in DSRCT. This study underscores the usefulness of preclinical in vitro models for studying molecular mechanisms and potential therapeutic options.
Collapse
Affiliation(s)
- Margit Bleijs
- Princess Máxima Center for Pediatric Oncology, 3584 CT Utrecht, The Netherlands; (M.B.); (C.P.); (S.E.); (F.R.); (F.M.-W.); (M.v.d.W.)
- Oncode Institute, 3521 AL Utrecht, The Netherlands
| | - Corine Pleijte
- Princess Máxima Center for Pediatric Oncology, 3584 CT Utrecht, The Netherlands; (M.B.); (C.P.); (S.E.); (F.R.); (F.M.-W.); (M.v.d.W.)
- Oncode Institute, 3521 AL Utrecht, The Netherlands
| | - Sem Engels
- Princess Máxima Center for Pediatric Oncology, 3584 CT Utrecht, The Netherlands; (M.B.); (C.P.); (S.E.); (F.R.); (F.M.-W.); (M.v.d.W.)
- Oncode Institute, 3521 AL Utrecht, The Netherlands
| | - Femke Ringnalda
- Princess Máxima Center for Pediatric Oncology, 3584 CT Utrecht, The Netherlands; (M.B.); (C.P.); (S.E.); (F.R.); (F.M.-W.); (M.v.d.W.)
- Oncode Institute, 3521 AL Utrecht, The Netherlands
| | - Friederike Meyer-Wentrup
- Princess Máxima Center for Pediatric Oncology, 3584 CT Utrecht, The Netherlands; (M.B.); (C.P.); (S.E.); (F.R.); (F.M.-W.); (M.v.d.W.)
| | - Marc van de Wetering
- Princess Máxima Center for Pediatric Oncology, 3584 CT Utrecht, The Netherlands; (M.B.); (C.P.); (S.E.); (F.R.); (F.M.-W.); (M.v.d.W.)
| | - Hans Clevers
- Princess Máxima Center for Pediatric Oncology, 3584 CT Utrecht, The Netherlands; (M.B.); (C.P.); (S.E.); (F.R.); (F.M.-W.); (M.v.d.W.)
- Oncode Institute, 3521 AL Utrecht, The Netherlands
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, 3584 CT Utrecht, The Netherlands
- Correspondence:
| |
Collapse
|
173
|
Lee S, Wang EY, Steinberg AB, Walton CC, Chinta SJ, Andersen JK. A guide to senolytic intervention in neurodegenerative disease. Mech Ageing Dev 2021; 200:111585. [PMID: 34627838 PMCID: PMC8627445 DOI: 10.1016/j.mad.2021.111585] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/20/2021] [Accepted: 10/06/2021] [Indexed: 12/18/2022]
Abstract
Cellular senescence is a potential tumor-suppressive mechanism that generally results in an irreversible cell cycle arrest. Senescent cells accumulate with age and actively secrete soluble factors, collectively termed the 'senescence-associated secretory phenotype' (SASP), which has both beneficial and detrimental effects. Although the contribution of senescent cells to age-related pathologies has been well-established outside the brain, emerging evidence indicates that brain cells also undergo cellular senescence and contribute to neuronal loss in the context of age-related neurodegenerative diseases. Contribution of senescent cells in the pathogenesis of neurological disorders has led to the possibility of eliminating senescence cells via pharmacological compounds called senolytics. Recently several senolytics have been demonstrated to elicit improved cognitive performance and healthspan in mouse models of neurodegeneration. However, their translation for use in the clinic still holds several potential challenges. This review summarizes available senolytics, their purported mode of action, and possible off-target effects. We also discuss possible alternative strategies that may help minimize potential side-effects associated with the senolytics approach.
Collapse
Affiliation(s)
- Suckwon Lee
- Buck Institute for Research on Aging, 8001 Redwood Blvd., Novato, CA, 94945, USA
| | - Ellen Y Wang
- Buck Institute for Research on Aging, 8001 Redwood Blvd., Novato, CA, 94945, USA
| | - Alexandra B Steinberg
- University of Wisconsin Department of Biochemistry, 433 Babcock Drive., Madison, WI, 53706, USA
| | - Chaska C Walton
- Buck Institute for Research on Aging, 8001 Redwood Blvd., Novato, CA, 94945, USA.
| | - Shankar J Chinta
- Buck Institute for Research on Aging, 8001 Redwood Blvd., Novato, CA, 94945, USA; Touro University California, College of Pharmacy, 1310 Club Dr., Vallejo, CA, 94592, USA.
| | - Julie K Andersen
- Buck Institute for Research on Aging, 8001 Redwood Blvd., Novato, CA, 94945, USA.
| |
Collapse
|
174
|
Siddiqui M, Konopleva M. Keeping up with venetoclax for leukemic malignancies: key findings, optimal regimens and clinical considerations. Expert Rev Clin Pharmacol 2021; 14:1497-1512. [PMID: 34791957 DOI: 10.1080/17512433.2021.2008239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Venetoclax has transformed the treatment landscape in hematologic malignancies, especially in elderly population. With high rates of remission, deep and durable responses, and safe toxicity profile, venetoclax in combination therapy has been extremely effective, garnering accelerated approval and becoming standard of care in lymphoid and myeloid malignancies. AREAS COVERED The role of venetoclax in the intrinsic apoptotic pathway is covered. This includes preclinical and clinical experience of venetoclax monotherapy and combination therapy in relapsed/refractory and frontline CLL, AML, ALL and high-risk MDS, with an emphasis on key clinical trials and efficacy of combination regimens in distinct mutational landscapes. Strategies to mitigate myelosuppression, manage dose adjustments and infectious complications are addressed. EXPERT OPINION Targeting BCL-2 offers a safe and highly effective adjunct to available therapies in hematologic malignancies. Despite success and frequent utilization of venetoclax, several resistance mechanisms have been elucidated, prompting development of novel combinatorial strategies. Further, on-target myelosuppression of venetoclax is a key obstacle in clinical practice, requiring diligent monitoring and practice-based knowledge of dose modifications. Despite these limitations, venetoclax has gained tremendous popularity in hematologic-oncology, becoming an integral component of numerous combination regimes, with ongoing plethora of clinical trials encompassing standard chemotherapy, targeted agents and immune-based approaches.
Collapse
Affiliation(s)
- Maria Siddiqui
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 428, Houston, TX, 77030, USA
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 428, Houston, TX, 77030, USA
| |
Collapse
|
175
|
Yamamoto M, Sanomachi T, Suzuki S, Togashi K, Sugai A, Seino S, Sato A, Okada M, Kitanaka C. Gemcitabine radiosensitization primes irradiated malignant meningioma cells for senolytic elimination by navitoclax. Neurooncol Adv 2021; 3:vdab148. [PMID: 34765973 PMCID: PMC8577526 DOI: 10.1093/noajnl/vdab148] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Background Malignant meningioma is an aggressive tumor that requires adjuvant radiotherapy after surgery, yet there has been no standard systemic therapy established so far. We recently reported that malignant meningioma cells are highly sensitive to gemcitabine; however, it remains unknown whether or how gemcitabine interacts with ionizing radiation (IR) in malignant meningioma cells. Methods We examined the radiosensitization effects of gemcitabine using malignant meningioma cell lines and xenografts and explored the underlying mechanisms. Results Gemcitabine sensitized malignant meningioma cells to IR through the induction of senescence both in vitro and in vivo. Gemcitabine augmented the intracellular production of reactive oxygen species (ROS) by IR, which, together with cell growth suppression/senescence induced by this combination, was inhibited by N-acetyl-cysteine, suggesting a pivotal role for ROS in these combinatorial effects. Navitoclax, a senolytic drug that inhibits Bcl-2 proteins, further enhanced the effects of the combination of gemcitabine and IR by strongly inducing apoptotic cell death in senescent cells. Conclusion These results not only indicate the potential of gemcitabine as a candidate radiosensitizer for malignant meningioma, but also reveal a novel role for gemcitabine radiosensitization as a means to create a therapeutic vulnerability of senescent meningioma cells to senolytics.
Collapse
Affiliation(s)
- Masahiro Yamamoto
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan
- Corresponding Author: Masahiro Yamamoto, MD, PhD, Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, 990-9585, Japan ()
| | - Tomomi Sanomachi
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan
- Department of Clinical Oncology, Yamagata University School of Medicine, Yamagata, Japan
| | - Shuhei Suzuki
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan
- Department of Clinical Oncology, Yamagata University School of Medicine, Yamagata, Japan
| | - Keita Togashi
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan
- Department of Ophthalmology and Visual Sciences, Yamagata University School of Medicine, Yamagata, Japan
| | - Asuka Sugai
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan
| | - Shizuka Seino
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan
| | - Atsushi Sato
- Department of Neurosurgery, Yamagata University School of Medicine, Yamagata, Japan
| | - Masashi Okada
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan
| | - Chifumi Kitanaka
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan
- Research Institute for Promotion of Medical Sciences, Yamagata University Faculty of Medicine, Yamagata, Japan
- Corresponding Author: Chifumi Kitanaka, MD, PhD, Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, 990-9585, Japan ()
| |
Collapse
|
176
|
Yadav M, Liu J, Song F, Mo X, Jacob NR, Xu-Welliver M, Chakravarti A, Jacob NK. Utility of circulating microRNA-150 for rapid evaluation of bone marrow depletion after radiation, and efficiency of bone marrow reconstitution. Int J Radiat Oncol Biol Phys 2021; 112:964-974. [PMID: 34767935 DOI: 10.1016/j.ijrobp.2021.10.150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Total body irradiation (TBI) is a common myeloablative preparative regimen used in acute myeloid and lymphoblastic leukemia patients prior to allogenic hematopoietic stem cell transplantation (HSCT). The inefficient clearance of tumor cells and radiation-induced toxicity to normal tissues is attributed to relapse and morbidity in a significant fraction of patients. Developing biomarkers that provide an individual's physiological response to radiation will allow personalized treatment and follow-up. We investigated the utility of circulating microRNA150-5p (miR150) for evaluation of radiation dose response. MATERIALS AND METHODS Age-, gender-, and strain-matched wild type and miR150 null (knock out, KO) mice were subjected to TBI and evaluated for the impact of circulating miR150 expression on survival and hematological endpoints. Dose- and time-dependent changes of the miR150 level in bone marrow were assessed using flow cytometry. The functional roles of miR150 in cellular response to radiation were evaluated using apoptosis assay. miR150 expression in leukemic cell lines and in blood collected from leukemia patients with diverse outcomes were evaluated by quantitative RT-PCR. RESULTS Absence of miR150 in mice conferred resistance to radiation injury and resulted in accelerated recovery of lymphoid and myeloid cells after ablative or partially ablative TBI in mice. Overexpression of miR150 resulted in a higher percentage of cells at G2/M phases of cell cycle which is associated with increased sensitivity and susceptibility to apoptotic cell death after radiation. Levels of circulating miR150 were found to be decreased after radiation in leukemia patients and exhibited an inverse correlation with recurrence. CONCLUSION Current study demonstrates the utility of a miR150-based blood test for rapid evaluation of the efficiency of marrow ablation and recovery following radiation and HSCT. The internally controlled blood test will potentially provide near real-time evaluation of functional marrow that will allow optimal dosing based on an individual's physiological response to radiation.
Collapse
Affiliation(s)
- Marshleen Yadav
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Joseph Liu
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Feifei Song
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Xiaokui Mo
- Center for Biostatistics, Ohio State University, Columbus, Ohio
| | - Nitya R Jacob
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Meng Xu-Welliver
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio; Department of Radiation Oncology, Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Arnab Chakravarti
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio; Department of Radiation Oncology, Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Naduparambil K Jacob
- Ohio State University Comprehensive Cancer Center, Columbus, Ohio; Department of Radiation Oncology, Ohio State University Wexner Medical Center, Columbus, Ohio.
| |
Collapse
|
177
|
Banerjee P, Kotla S, Reddy Velatooru L, Abe RJ, Davis EA, Cooke JP, Schadler K, Deswal A, Herrmann J, Lin SH, Abe JI, Le NT. Senescence-Associated Secretory Phenotype as a Hinge Between Cardiovascular Diseases and Cancer. Front Cardiovasc Med 2021; 8:763930. [PMID: 34746270 PMCID: PMC8563837 DOI: 10.3389/fcvm.2021.763930] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 09/16/2021] [Indexed: 12/13/2022] Open
Abstract
Overlapping risks for cancer and cardiovascular diseases (CVD), the two leading causes of mortality worldwide, suggest a shared biology between these diseases. The role of senescence in the development of cancer and CVD has been established. However, its role as the intersection between these diseases remains unclear. Senescence was originally characterized by an irreversible cell cycle arrest after a high number of divisions, namely replicative senescence (RS). However, it is becoming clear that senescence can also be instigated by cellular stress, so-called stress-induced premature senescence (SIPS). Telomere shortening is a hallmark of RS. The contribution of telomere DNA damage and subsequent DNA damage response/repair to SIPS has also been suggested. Although cellular senescence can mediate cell cycle arrest, senescent cells can also remain metabolically active and secrete cytokines, chemokines, growth factors, and reactive oxygen species (ROS), so-called senescence-associated secretory phenotype (SASP). The involvement of SASP in both cancer and CVD has been established. In patients with cancer or CVD, SASP is induced by various stressors including cancer treatments, pro-inflammatory cytokines, and ROS. Therefore, SASP can be the intersection between cancer and CVD. Importantly, the conventional concept of senescence as the mediator of cell cycle arrest has been challenged, as it was recently reported that chemotherapy-induced senescence can reprogram senescent cancer cells to acquire “stemness” (SAS: senescence-associated stemness). SAS allows senescent cancer cells to escape cell cycle arrest with strongly enhanced clonogenic growth capacity. SAS supports senescent cells to promote both cancer and CVD, particularly in highly stressful conditions such as cancer treatments, myocardial infarction, and heart failure. As therapeutic advances have increased overlapping risk factors for cancer and CVD, to further understand their interaction may provide better prevention, earlier detection, and safer treatment. Thus, it is critical to study the mechanisms by which these senescence pathways (SAS/SASP) are induced and regulated in both cancer and CVD.
Collapse
Affiliation(s)
- Priyanka Banerjee
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Loka Reddy Velatooru
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Rei J Abe
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Elizabeth A Davis
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - John P Cooke
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Keri Schadler
- Department of Pediatrics Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Anita Deswal
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Joerg Herrmann
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Steven H Lin
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jun-Ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nhat-Tu Le
- Center for Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
178
|
Townsend PA, Kozhevnikova MV, Cexus ONF, Zamyatnin AA, Soond SM. BH3-mimetics: recent developments in cancer therapy. J Exp Clin Cancer Res 2021; 40:355. [PMID: 34753495 PMCID: PMC8576916 DOI: 10.1186/s13046-021-02157-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/26/2021] [Indexed: 01/11/2023] Open
Abstract
The hopeful outcomes from 30 years of research in BH3-mimetics have indeed served a number of solid paradigms for targeting intermediates from the apoptosis pathway in a variety of diseased states. Not only have such rational approaches in drug design yielded several key therapeutics, such outputs have also offered insights into the integrated mechanistic aspects of basic and clinical research at the genetics level for the future. In no other area of medical research have the effects of such work been felt, than in cancer research, through targeting the BAX-Bcl-2 protein-protein interactions. With these promising outputs in mind, several mimetics, and their potential therapeutic applications, have also been developed for several other pathological conditions, such as cardiovascular disease and tissue fibrosis, thus highlighting the universal importance of the intrinsic arm of the apoptosis pathway and its input to general tissue homeostasis. Considering such recent developments, and in a field that has generated so much scientific interest, we take stock of how the broadening area of BH3-mimetics has developed and diversified, with a focus on their uses in single and combined cancer treatment regimens and recently explored therapeutic delivery methods that may aid the development of future therapeutics of this nature.
Collapse
Affiliation(s)
- Paul A Townsend
- University of Surrey, Guildford, UK.
- Sechenov First Moscow State Medical University, Moscow, Russian Federation.
- University of Manchester, Manchester, UK.
| | - Maria V Kozhevnikova
- University of Surrey, Guildford, UK
- Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | | | - Andrey A Zamyatnin
- University of Surrey, Guildford, UK
- Sechenov First Moscow State Medical University, Moscow, Russian Federation
- Lomonosov Moscow State University, Moscow, Russian Federation
- Sirius University of Science and Technology, Sochi, Russian Federation
| | - Surinder M Soond
- University of Surrey, Guildford, UK.
- Sechenov First Moscow State Medical University, Moscow, Russian Federation.
| |
Collapse
|
179
|
Venetoclax in Acute Myeloid Leukemia: Molecular Basis, Evidences for Preclinical and Clinical Efficacy and Strategies to Target Resistance. Cancers (Basel) 2021; 13:cancers13225608. [PMID: 34830763 PMCID: PMC8615921 DOI: 10.3390/cancers13225608] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 10/23/2021] [Accepted: 11/03/2021] [Indexed: 12/27/2022] Open
Abstract
Venetoclax is a BH3-mimetics agent specifically interacting with the antiapoptotic protein BCL-2, facilitating cytochrome c release from mitochondria, subsequent caspases activation, and cell death. Utilization of venetoclax has profoundly changed the landscape of treatment for the poor-prognosis category of AML patients unfit for intensive chemotherapy. In the phase III VIALE-A study, Venetoclax, in combination with the hypomethylating agent azacitidine, showed a 65% overall response rate and 14.7-month overall survival, in comparison with 22% and 8 months in the control arm. These results led to the widespread use of venetoclax in this indication. Other combination regimens, consisting of low-intensity, intensive, or targeted therapies are currently under evaluation. Despite promising results, preventing relapses or resistance to venetoclax is still an unmet clinical need. Numerous studies have been conducted to identify and overcome venetoclax resistance in preclinical models or in clinical trials, including the inhibition of other antiapoptotic proteins, the induction of proapoptotic BH3-only proteins, and/or the targeting of the mitochondrial metabolism and machinery.
Collapse
|
180
|
Lim S, Kim TJ, Kim YJ, Kim C, Ko SB, Kim BS. Senolytic Therapy for Cerebral Ischemia-Reperfusion Injury. Int J Mol Sci 2021; 22:ijms222111967. [PMID: 34769397 PMCID: PMC8584561 DOI: 10.3390/ijms222111967] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 10/30/2021] [Accepted: 11/02/2021] [Indexed: 12/13/2022] Open
Abstract
Ischemic stroke is one of the leading causes of death, and even timely treatment can result in severe disabilities. Reperfusion of the ischemic stroke region and restoration of the blood supply often lead to a series of cellular and biochemical consequences, including generation of reactive oxygen species (ROS), expression of inflammatory cytokines, inflammation, and cerebral cell damage, which is collectively called cerebral ischemia-reperfusion (IR) injury. Since ROS and inflammatory cytokines are involved in cerebral IR injury, injury could involve cellular senescence. Thus, we investigated whether senolytic therapy that eliminates senescent cells could be an effective treatment for cerebral IR injury. To determine whether IR induces neural cell senescence in vitro, astrocytes were subjected to oxygen-glucose deprivation/reoxygenation (OGD/R). OGD/R induced astrocyte senescence and senescent cells in OGD/R-injured astrocytes were effectively eliminated in vitro by ABT263, a senolytic agent. IR in rats with intraluminal middle cerebral artery occlusion induced cellular senescence in the ischemic region. The senescent cells in IR-injured rats were effectively eliminated by intravenous injections of ABT263. Importantly, ABT263 treatment significantly reduced the infarct volume and improved neurological function in behavioral tests. This study demonstrated, for the first time, that senolytic therapy has therapeutic potential for cerebral IR injury.
Collapse
Affiliation(s)
- Songhyun Lim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Korea; (S.L.); (C.K.)
| | - Tae Jung Kim
- Department of Neurology, Seoul National University Hospital, Seoul 03080, Korea; (T.J.K.); (Y.-J.K.)
- Department of Critical Care Medicine, Seoul National University Hospital, Seoul 03080, Korea
| | - Young-Ju Kim
- Department of Neurology, Seoul National University Hospital, Seoul 03080, Korea; (T.J.K.); (Y.-J.K.)
| | - Cheesue Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Korea; (S.L.); (C.K.)
| | - Sang-Bae Ko
- Department of Neurology, Seoul National University Hospital, Seoul 03080, Korea; (T.J.K.); (Y.-J.K.)
- Department of Critical Care Medicine, Seoul National University Hospital, Seoul 03080, Korea
- Correspondence: (S.-B.K.); (B.-S.K.); Tel.: +82-2-2072-2278 (S.-B.K.); +82-2-880-1509 (B.-S.K.)
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Korea; (S.L.); (C.K.)
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul 08826, Korea
- Institute of Chemical Processes, Seoul National University, Seoul 08826, Korea
- Institute of Engineering Research, Seoul National University, Seoul 08826, Korea
- Bio-MAX Institute, Seoul National University, Seoul 08826, Korea
- Correspondence: (S.-B.K.); (B.-S.K.); Tel.: +82-2-2072-2278 (S.-B.K.); +82-2-880-1509 (B.-S.K.)
| |
Collapse
|
181
|
Lee EF, Fairlie WD. Discovery, development and application of drugs targeting BCL-2 pro-survival proteins in cancer. Biochem Soc Trans 2021; 49:2381-2395. [PMID: 34515749 PMCID: PMC8589430 DOI: 10.1042/bst20210749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022]
Abstract
The discovery of a new class of small molecule compounds that target the BCL-2 family of anti-apoptotic proteins is one of the great success stories of basic science leading to translational outcomes in the last 30 years. The eponymous BCL-2 protein was identified over 30 years ago due to its association with cancer. However, it was the unveiling of the biochemistry and structural biology behind it and its close relatives' mechanism(s)-of-action that provided the inspiration for what are now known as 'BH3-mimetics', the first clinically approved drugs designed to specifically inhibit protein-protein interactions. Herein, we chart the history of how these drugs were discovered, their evolution and application in cancer treatment.
Collapse
Affiliation(s)
- Erinna F. Lee
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
- Cell Death and Survival Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria 3086, Australia
| | - W. Douglas Fairlie
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
- Cell Death and Survival Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria 3084, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria 3086, Australia
| |
Collapse
|
182
|
Lee JR, Park BW, Park JH, Lim S, Kwon SP, Hwang JW, Kim H, Park HJ, Kim BS. Local delivery of a senolytic drug in ischemia and reperfusion-injured heart attenuates cardiac remodeling and restores impaired cardiac function. Acta Biomater 2021; 135:520-533. [PMID: 34454081 DOI: 10.1016/j.actbio.2021.08.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 12/15/2022]
Abstract
Myocardial ischemia-reperfusion (IR) generates stress-induced senescent cells (SISCs) that play an important role in the pathophysiology of adverse cardiac remodeling and heart failure via secretion of pro-inflammatory molecules and matrix-degrading proteases. Thus, removal of senescent cells using a senolytic drug could be a potentially effective treatment. However, clinical studies on cancer treatment with a senolytic drug have revealed that systemic administration of a senolytic drug often causes systemic toxicity. Herein we show for the first time that local delivery of a senolytic drug can effectively treat myocardial IR injury. We found that biodegradable poly(lactic-co-glycolic acid) nanoparticle-based local delivery of a senolytic drug (ABT263-PLGA) successfully eliminated SISCs in the IR-injured rat hearts without systemic toxicity. Consequently, the treatment ameliorated inflammatory responses and attenuated adverse remodeling. Surprisingly, the ABT263-PLGA treatment restored the cardiac function over time, whereas the cardiac function decreased over time in the no treatment group. Mechanistically, the ABT263-PLGA treatment not only markedly reduced the expression of pro-inflammatory molecules and matrix-degrading proteases, but also induced macrophage polarization from the inflammatory phase to the reparative phase via efferocytosis of apoptotic SISCs by macrophages. Therefore, the senolytic strategy with ABT263-PLGA in the early stage of myocardial IR injury may be an effective therapeutic option for myocardial infarction. STATEMENT OF SIGNIFICANCE: This study describes a local injection of senolytic drug-loaded nanoparticles that selectively kills stress-induced senescent cells (SISCs) in infarcted heart. Removal of SISCs decreases inflammatory cytokines and normal cell death. We firstly revealed that further efferocytosis of apoptotic senescent cells by macrophages restores cardiac function after myocardial ischemia-reperfusion injury. Importantly, a local injection of senolytic drug did not exhibit systemic toxicity, but a systemic injection did. Our findings not only spotlight the basic understanding of therapeutic potential of senolysis in infarcted myocardium, but also pave the way for the further application of senolytic drug for non-aging related diseases.
Collapse
Affiliation(s)
- Ju-Ro Lee
- Chemical and Biological Engineering, Seoul National University, Seoul 08826, South Korea
| | - Bong-Woo Park
- Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Division of Cardiology, Department of Internal Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Jae-Hyun Park
- Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Division of Cardiology, Department of Internal Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Songhyun Lim
- Chemical and Biological Engineering, Seoul National University, Seoul 08826, South Korea
| | - Sung Pil Kwon
- Chemical and Biological Engineering, Seoul National University, Seoul 08826, South Korea
| | - Ji-Won Hwang
- Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Division of Cardiology, Department of Internal Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Hyeok Kim
- Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Division of Cardiology, Department of Internal Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Hun-Jun Park
- Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Division of Cardiology, Department of Internal Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, Seoul 06591, South Korea.
| | - Byung-Soo Kim
- Chemical and Biological Engineering, Seoul National University, Seoul 08826, South Korea; Institute of Chemical Processes, Institute of Engineering Research, Seoul National University, Seoul 08826, South Korea.
| |
Collapse
|
183
|
Miller KN, Victorelli SG, Salmonowicz H, Dasgupta N, Liu T, Passos JF, Adams PD. Cytoplasmic DNA: sources, sensing, and role in aging and disease. Cell 2021; 184:5506-5526. [PMID: 34715021 PMCID: PMC8627867 DOI: 10.1016/j.cell.2021.09.034] [Citation(s) in RCA: 162] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 12/17/2022]
Abstract
Endogenous cytoplasmic DNA (cytoDNA) species are emerging as key mediators of inflammation in diverse physiological and pathological contexts. Although the role of endogenous cytoDNA in innate immune activation is well established, the cytoDNA species themselves are often poorly characterized and difficult to distinguish, and their mechanisms of formation, scope of function and contribution to disease are incompletely understood. Here, we summarize current knowledge in this rapidly progressing field with emphases on similarities and differences between distinct cytoDNAs, their underlying molecular mechanisms of formation and function, interactions between cytoDNA pathways, and therapeutic opportunities in the treatment of age-associated diseases.
Collapse
Affiliation(s)
- Karl N Miller
- Aging, Cancer and Immuno-oncology Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Stella G Victorelli
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Hanna Salmonowicz
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA; Institute for Cell and Molecular Biosciences & Newcastle University Institute for Ageing, Newcastle upon Tyne NE4 5PL, UK; International Institute of Molecular Mechanisms and Machines, Polish Academy of Sciences, 02-109 Warsaw, Poland
| | - Nirmalya Dasgupta
- Aging, Cancer and Immuno-oncology Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Tianhui Liu
- Aging, Cancer and Immuno-oncology Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - João F Passos
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA.
| | - Peter D Adams
- Aging, Cancer and Immuno-oncology Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
184
|
Paysant H, Hedir S, Justaud F, Weiswald LB, El Dine AN, Soulieman A, Hachem A, Elie N, Brotin E, Denoyelle C, Bignon J, Roussi F, Jouanne M, Tasseau O, Roisnel T, Voisin-Chiret AS, Grée R, Levoin N, Poulain L. Structural revision of the Mcl-1 inhibitor MIM1: synthesis and biological studies on ovarian cancer cells with evaluation of designed analogues. Org Biomol Chem 2021; 19:8968-8987. [PMID: 34596646 DOI: 10.1039/d1ob01521d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the area of cancer research, the development of new and potent inhibitors of anti-apoptotic proteins is a very active and promising topic. The small molecule MIM1 has been reported earlier as one of the first selective inhibitors of the anti-apoptotic protein Mcl-1. In the present paper, we first revised the structure of this molecule based on extensive physicochemical analyses. Then we designed and synthesized a focused library of analogues for the corrected structure of MIM1. Next, these molecules were subjected to a panel of in cellulo biological studies, allowing the identification of dual Bcl-xL/Mcl-1 inhibitors, as well as selective Mcl-1 inhibitors. These results have been complemented by fluorescence polarization assays with the Mcl-1 protein. Preliminary structure-activity relationships were discussed and extensive molecular modelling studies allowed us to propose a rationale for the biological activity of this series of new inhibitors, in particular for the selectivity of inhibition of Mcl-1 versus Bcl-xL.
Collapse
Affiliation(s)
- Hippolyte Paysant
- Normandie Univ, UNICAEN, Inserm U1086 ANTICIPE "Unité de Recherche Interdisciplinaire pour la Prévention et le Traitement des Cancers", Caen, France
- UNICANCER, Centre de Lutte Contre le Cancer F. Baclesse, 3 avenue du Général Harris, 14076, Caen, France
| | - Siham Hedir
- Normandie Univ, UNICAEN, Inserm U1086 ANTICIPE "Unité de Recherche Interdisciplinaire pour la Prévention et le Traitement des Cancers", Caen, France
- UNICANCER, Centre de Lutte Contre le Cancer F. Baclesse, 3 avenue du Général Harris, 14076, Caen, France
| | - Frédéric Justaud
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes), UMR 6226, F-35000, Rennes, France.
| | - Louis Bastien Weiswald
- Normandie Univ, UNICAEN, Inserm U1086 ANTICIPE "Unité de Recherche Interdisciplinaire pour la Prévention et le Traitement des Cancers", Caen, France
- UNICANCER, Centre de Lutte Contre le Cancer F. Baclesse, 3 avenue du Général Harris, 14076, Caen, France
| | - Assaad Nasr El Dine
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes), UMR 6226, F-35000, Rennes, France.
- Laboratoire de Chimie Médicinale et de Produits Naturels, Université Libanaise, Faculté des Sciences et PRASE-EDST, Hadath, Beyrouth, Liban
| | - Ali Soulieman
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes), UMR 6226, F-35000, Rennes, France.
- Laboratoire de Chimie Médicinale et de Produits Naturels, Université Libanaise, Faculté des Sciences et PRASE-EDST, Hadath, Beyrouth, Liban
| | - Ali Hachem
- Laboratoire de Chimie Médicinale et de Produits Naturels, Université Libanaise, Faculté des Sciences et PRASE-EDST, Hadath, Beyrouth, Liban
| | - Nicolas Elie
- Normandie Univ, UNICAEN, SF 4206 ICORE, CMABIO3, Caen, France
| | - Emilie Brotin
- Normandie Univ, UNICAEN, Inserm U1086 ANTICIPE "Unité de Recherche Interdisciplinaire pour la Prévention et le Traitement des Cancers", Caen, France
- UNICANCER, Centre de Lutte Contre le Cancer F. Baclesse, 3 avenue du Général Harris, 14076, Caen, France
- Normandie Univ, UNICAEN, SF 4206 ICORE, CMABIO3, Caen, France
- Normandie Univ, UNICAEN, SF4206 ICORE, Plateforme ImpedanCELL, Caen, France
| | - Christophe Denoyelle
- Normandie Univ, UNICAEN, Inserm U1086 ANTICIPE "Unité de Recherche Interdisciplinaire pour la Prévention et le Traitement des Cancers", Caen, France
- UNICANCER, Centre de Lutte Contre le Cancer F. Baclesse, 3 avenue du Général Harris, 14076, Caen, France
- Normandie Univ, UNICAEN, SF 4206 ICORE, CMABIO3, Caen, France
- Normandie Univ, UNICAEN, SF4206 ICORE, Plateforme ImpedanCELL, Caen, France
| | - Jérôme Bignon
- Institut de Chimie des Substances Naturelles CNRS UPR 2301, Université Paris Saclay, Gif-sur-Yvette, France
| | - Fanny Roussi
- Institut de Chimie des Substances Naturelles CNRS UPR 2301, Université Paris Saclay, Gif-sur-Yvette, France
| | - Marie Jouanne
- Normandie Univ, UNICAEN, Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), 14000 Caen, France
| | - Olivier Tasseau
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes), UMR 6226, F-35000, Rennes, France.
| | - Thierry Roisnel
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes), UMR 6226, F-35000, Rennes, France.
| | - Anne Sophie Voisin-Chiret
- Normandie Univ, UNICAEN, Centre d'Etudes et de Recherche sur le Médicament de Normandie (CERMN), 14000 Caen, France
| | - René Grée
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes), UMR 6226, F-35000, Rennes, France.
| | - Nicolas Levoin
- Bioprojet-Biotech, 4 rue du Chesnay Beauregard, BP 96205, 35762, Saint Grégoire, France
| | - Laurent Poulain
- Normandie Univ, UNICAEN, Inserm U1086 ANTICIPE "Unité de Recherche Interdisciplinaire pour la Prévention et le Traitement des Cancers", Caen, France
- UNICANCER, Centre de Lutte Contre le Cancer F. Baclesse, 3 avenue du Général Harris, 14076, Caen, France
| |
Collapse
|
185
|
Donati G, Ravà M, Filipuzzi M, Nicoli P, Cassina L, Verrecchia A, Doni M, Rodighiero S, Parodi F, Boletta A, Vellano CP, Marszalek JR, Draetta GF, Amati B. Targeting mitochondrial respiration and the BCL2 family in high-grade MYC-associated B-cell lymphoma. Mol Oncol 2021; 16:1132-1152. [PMID: 34632715 PMCID: PMC8895457 DOI: 10.1002/1878-0261.13115] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/27/2021] [Accepted: 10/08/2021] [Indexed: 01/02/2023] Open
Abstract
Multiple molecular features, such as activation of specific oncogenes (e.g., MYC, BCL2) or a variety of gene expression signatures, have been associated with disease course in diffuse large B‐cell lymphoma (DLBCL), although their relationships and implications for targeted therapy remain to be fully unraveled. We report that MYC activity is closely correlated with—and most likely a driver of—gene signatures related to oxidative phosphorylation (OxPhos) in DLBCL, pointing to OxPhos enzymes, in particular mitochondrial electron transport chain (ETC) complexes, as possible therapeutic targets in high‐grade MYC‐associated lymphomas. In our experiments, indeed, MYC sensitized B cells to the ETC complex I inhibitor IACS‐010759. Mechanistically, IACS‐010759 triggered the integrated stress response (ISR) pathway, driven by the transcription factors ATF4 and CHOP, which engaged the intrinsic apoptosis pathway and lowered the apoptotic threshold in MYC‐overexpressing cells. In line with these findings, the BCL2‐inhibitory compound venetoclax synergized with IACS‐010759 against double‐hit lymphoma (DHL), a high‐grade malignancy with concurrent activation of MYC and BCL2. In BCL2‐negative lymphoma cells, instead, killing by IACS‐010759 was potentiated by the Mcl‐1 inhibitor S63845. Thus, combining an OxPhos inhibitor with select BH3‐mimetic drugs provides a novel therapeutic principle against aggressive, MYC‐associated DLBCL variants.
Collapse
Affiliation(s)
- Giulio Donati
- European Institute of Oncology (IEO)-IRCCS, Milan, Italy
| | - Micol Ravà
- European Institute of Oncology (IEO)-IRCCS, Milan, Italy
| | | | - Paola Nicoli
- European Institute of Oncology (IEO)-IRCCS, Milan, Italy
| | - Laura Cassina
- IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Mirko Doni
- European Institute of Oncology (IEO)-IRCCS, Milan, Italy
| | | | | | | | - Christopher P Vellano
- Translational Research to Advance Therapeutics and Innovation in Oncology (TRACTION), Houston, TX, USA
| | - Joseph R Marszalek
- Translational Research to Advance Therapeutics and Innovation in Oncology (TRACTION), Houston, TX, USA
| | - Giulio F Draetta
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bruno Amati
- European Institute of Oncology (IEO)-IRCCS, Milan, Italy
| |
Collapse
|
186
|
Tarantini S, Balasubramanian P, Delfavero J, Csipo T, Yabluchanskiy A, Kiss T, Nyúl-Tóth Á, Mukli P, Toth P, Ahire C, Ungvari A, Benyo Z, Csiszar A, Ungvari Z. Treatment with the BCL-2/BCL-xL inhibitor senolytic drug ABT263/Navitoclax improves functional hyperemia in aged mice. GeroScience 2021; 43:2427-2440. [PMID: 34427858 PMCID: PMC8599595 DOI: 10.1007/s11357-021-00440-z] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/11/2021] [Indexed: 12/13/2022] Open
Abstract
Moment-to-moment adjustment of regional cerebral blood flow to neuronal activity via neurovascular coupling (NVC or "functional hyperemia") has a critical role in maintenance of healthy cognitive function. Aging-induced impairment of NVC responses importantly contributes to age-related cognitive decline. Advanced aging is associated with increased prevalence of senescent cells in the cerebral microcirculation, but their role in impaired NVC responses remains unexplored. The present study was designed to test the hypothesis that a validated senolytic treatment can improve NVC responses and cognitive performance in aged mice. To achieve this goal, aged (24-month-old) C57BL/6 mice were treated with ABT263/Navitoclax, a potent senolytic agent known to eliminate senescent cells in the aged mouse brain. Mice were behaviorally evaluated (radial arms water maze) and NVC was assessed by measuring CBF responses (laser speckle contrast imaging) in the somatosensory whisker barrel cortex evoked by contralateral whisker stimulation. We found that NVC responses were significantly impaired in aged mice. ABT263/Navitoclax treatment improved NVC response, which was associated with significantly improved hippocampal-encoded functions of learning and memory. ABT263/Navitoclax treatment did not significantly affect endothelium-dependent acetylcholine-induced relaxation of aorta rings. Thus, increased presence of senescent cells in the aged brain likely contributes to age-related neurovascular uncoupling, exacerbating cognitive decline. The neurovascular protective effects of ABT263/Navitoclax treatment highlight the preventive and therapeutic potential of senolytic treatments (as monotherapy or as part of combination treatment regimens) as effective interventions in patients at risk for vascular cognitive impairment (VCI).
Collapse
Affiliation(s)
- Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA.
- Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK, 73104, USA.
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary.
| | - Priya Balasubramanian
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
| | - Jordan Delfavero
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
| | - Tamas Csipo
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
- Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK, 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tamas Kiss
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
- Department of Pediatrics, University of Szeged, Szeged, Hungary
- First Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Ádám Nyúl-Tóth
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Szeged, Hungary
| | - Peter Mukli
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Peter Toth
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Neurosurgery, University of Pécs Clinical Center, Pecs, Hungary
- MTA-PTE Clinical Neuroscience MR Research Group, Pecs, Hungary
| | - Chetan Ahire
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
| | - Anna Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
| | - Zoltan Benyo
- International Training Program in Geroscience/Vascular Cognitive Impairment and Neurodegeneration Program, Department of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
- Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience/Vascular Cognitive Impairment and Neurodegeneration Program, Department of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 731042, USA
- Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK, 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| |
Collapse
|
187
|
New insights into binding of natural chalcones to Bcl-2, Bcl-xL and Mcl-1 anti-apoptotic proteins. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
188
|
Gasek NS, Kuchel GA, Kirkland JL, Xu M. Strategies for Targeting Senescent Cells in Human Disease. NATURE AGING 2021; 1:870-879. [PMID: 34841261 PMCID: PMC8612694 DOI: 10.1038/s43587-021-00121-8] [Citation(s) in RCA: 289] [Impact Index Per Article: 72.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 08/27/2021] [Indexed: 12/26/2022]
Abstract
Cellular senescence represents a distinct cell fate characterized by replicative arrest in response to a host of extrinsic and intrinsic stresses. Senescence provides programming during development and wound healing, while limiting tumorigenesis. However, pathologic accumulation of senescent cells is implicated in a range of diseases and age-associated morbidities across organ systems. Senescent cells produce distinct paracrine and endocrine signals, causing local tissue dysfunction and exerting deleterious systemic effects. Senescent cell removal by apoptosis-inducing "senolytic" agents or therapies that inhibit the senescence-associated secretory phenotype, SASP inhibitors, have demonstrated benefit in both pre-clinical and clinical models of geriatric decline and chronic diseases, suggesting senescent cells represent a pharmacologic target for alleviating effects of fundamental aging processes. However, senescent cell populations are heterogeneous in form, function, tissue distribution, and even differ among species, possibly explaining issues of bench-to-bedside translation in current clinical trials. Here, we review features of senescent cells and strategies for targeting them, including immunologic approaches, as well as key intracellular signaling pathways. Additionally, we survey current senolytic therapies in human trials. Collectively, there is demand for research to develop targeted senotherapeutics that address the needs of the aging and chronically-ill.
Collapse
Affiliation(s)
- Nathan S. Gasek
- UConn Center on Aging, UConn Health, Farmington, CT
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT
| | | | | | - Ming Xu
- UConn Center on Aging, UConn Health, Farmington, CT
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT
| |
Collapse
|
189
|
Gorombei P, Guidez F, Ganesan S, Chiquet M, Pellagatti A, Goursaud L, Tekin N, Beurlet S, Patel S, Guerenne L, Le Pogam C, Setterblad N, de la Grange P, LeBoeuf C, Janin A, Noguera ME, Sarda-Mantel L, Merlet P, Boultwood J, Konopleva M, Andreeff M, West R, Pla M, Adès L, Fenaux P, Krief P, Chomienne C, Omidvar N, Padua RA. BCL-2 Inhibitor ABT-737 Effectively Targets Leukemia-Initiating Cells with Differential Regulation of Relevant Genes Leading to Extended Survival in a NRAS/BCL-2 Mouse Model of High Risk-Myelodysplastic Syndrome. Int J Mol Sci 2021; 22:ijms221910658. [PMID: 34638998 PMCID: PMC8508829 DOI: 10.3390/ijms221910658] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 11/16/2022] Open
Abstract
During transformation, myelodysplastic syndromes (MDS) are characterized by reducing apoptosis of bone marrow (BM) precursors. Mouse models of high risk (HR)-MDS and acute myelogenous leukemia (AML) post-MDS using mutant NRAS and overexpression of human BCL-2, known to be poor prognostic indicators of the human diseases, were created. We have reported the efficacy of the BCL-2 inhibitor, ABT-737, on the AML post-MDS model; here, we report that this BCL-2 inhibitor also significantly extended survival of the HR-MDS mouse model, with reductions of BM blasts and lineage negative/Sca1+/KIT+ (LSK) cells. Secondary transplants showed increased survival in treated compared to untreated mice. Unlike the AML model, BCL-2 expression and RAS activity decreased following treatment and the RAS:BCL-2 complex remained in the plasma membrane. Exon-specific gene expression profiling (GEP) of HR-MDS mice showed 1952 differentially regulated genes upon treatment, including genes important for the regulation of stem cells, differentiation, proliferation, oxidative phosphorylation, mitochondrial function, and apoptosis; relevant in human disease. Spliceosome genes, found to be abnormal in MDS patients and downregulated in our HR-MDS model, such as Rsrc1 and Wbp4, were upregulated by the treatment, as were genes involved in epigenetic regulation, such as DNMT3A and B, upregulated upon disease progression and downregulated upon treatment.
Collapse
Affiliation(s)
- Petra Gorombei
- INSERM UMR-S1131, Université de Paris, Institut de la Recherche Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis Hôpital, 75010 Paris, France; (P.G.); (F.G.); (S.G.); (M.C.); (L.G.); (N.T.); (S.B.); (S.P.); (L.G.); (C.L.P.); (M.P.); (P.K.); (C.C.)
| | - Fabien Guidez
- INSERM UMR-S1131, Université de Paris, Institut de la Recherche Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis Hôpital, 75010 Paris, France; (P.G.); (F.G.); (S.G.); (M.C.); (L.G.); (N.T.); (S.B.); (S.P.); (L.G.); (C.L.P.); (M.P.); (P.K.); (C.C.)
| | - Saravanan Ganesan
- INSERM UMR-S1131, Université de Paris, Institut de la Recherche Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis Hôpital, 75010 Paris, France; (P.G.); (F.G.); (S.G.); (M.C.); (L.G.); (N.T.); (S.B.); (S.P.); (L.G.); (C.L.P.); (M.P.); (P.K.); (C.C.)
| | - Mathieu Chiquet
- INSERM UMR-S1131, Université de Paris, Institut de la Recherche Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis Hôpital, 75010 Paris, France; (P.G.); (F.G.); (S.G.); (M.C.); (L.G.); (N.T.); (S.B.); (S.P.); (L.G.); (C.L.P.); (M.P.); (P.K.); (C.C.)
| | - Andrea Pellagatti
- Blood Cancer UK Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and BRC Haematology Theme, Oxford OX3 9DU, UK; (A.P.); (J.B.)
| | - Laure Goursaud
- INSERM UMR-S1131, Université de Paris, Institut de la Recherche Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis Hôpital, 75010 Paris, France; (P.G.); (F.G.); (S.G.); (M.C.); (L.G.); (N.T.); (S.B.); (S.P.); (L.G.); (C.L.P.); (M.P.); (P.K.); (C.C.)
| | - Nilgun Tekin
- INSERM UMR-S1131, Université de Paris, Institut de la Recherche Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis Hôpital, 75010 Paris, France; (P.G.); (F.G.); (S.G.); (M.C.); (L.G.); (N.T.); (S.B.); (S.P.); (L.G.); (C.L.P.); (M.P.); (P.K.); (C.C.)
| | - Stephanie Beurlet
- INSERM UMR-S1131, Université de Paris, Institut de la Recherche Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis Hôpital, 75010 Paris, France; (P.G.); (F.G.); (S.G.); (M.C.); (L.G.); (N.T.); (S.B.); (S.P.); (L.G.); (C.L.P.); (M.P.); (P.K.); (C.C.)
| | - Satyananda Patel
- INSERM UMR-S1131, Université de Paris, Institut de la Recherche Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis Hôpital, 75010 Paris, France; (P.G.); (F.G.); (S.G.); (M.C.); (L.G.); (N.T.); (S.B.); (S.P.); (L.G.); (C.L.P.); (M.P.); (P.K.); (C.C.)
| | - Laura Guerenne
- INSERM UMR-S1131, Université de Paris, Institut de la Recherche Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis Hôpital, 75010 Paris, France; (P.G.); (F.G.); (S.G.); (M.C.); (L.G.); (N.T.); (S.B.); (S.P.); (L.G.); (C.L.P.); (M.P.); (P.K.); (C.C.)
| | - Carole Le Pogam
- INSERM UMR-S1131, Université de Paris, Institut de la Recherche Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis Hôpital, 75010 Paris, France; (P.G.); (F.G.); (S.G.); (M.C.); (L.G.); (N.T.); (S.B.); (S.P.); (L.G.); (C.L.P.); (M.P.); (P.K.); (C.C.)
| | - Niclas Setterblad
- Imagerie Département, Université de Paris, Institut de la Recherche Saint-Louis, 75010 Paris, France;
| | - Pierre de la Grange
- GenoSplice Technology, Paris Biotech Santé, 29 Rue du Faubourg Saint-Jacques, 75014 Paris, France;
| | - Christophe LeBoeuf
- INSERM UMR-S942, Université de Paris, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis, 75010 Paris, France; (C.L.); (A.J.)
| | - Anne Janin
- INSERM UMR-S942, Université de Paris, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis, 75010 Paris, France; (C.L.); (A.J.)
| | - Maria-Elena Noguera
- Department of Cytology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis, 75010 Paris, France;
| | - Laure Sarda-Mantel
- Radiopharmacie AP-HP, Hôpital Saint-Louis, Service Medicine Nuclear, AP-HP Lariboisiere, 75010 Paris, France;
| | - Pascale Merlet
- Nuclear Medicine, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis, 75010 Paris, France;
| | - Jacqueline Boultwood
- Blood Cancer UK Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, and BRC Haematology Theme, Oxford OX3 9DU, UK; (A.P.); (J.B.)
| | - Marina Konopleva
- M. D. Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA; (M.K.); (M.A.)
| | - Michael Andreeff
- M. D. Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA; (M.K.); (M.A.)
| | - Robert West
- Department of Public Health, Cardiff University School of Medicine, Cardiff CF14 4XN, UK;
| | - Marika Pla
- INSERM UMR-S1131, Université de Paris, Institut de la Recherche Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis Hôpital, 75010 Paris, France; (P.G.); (F.G.); (S.G.); (M.C.); (L.G.); (N.T.); (S.B.); (S.P.); (L.G.); (C.L.P.); (M.P.); (P.K.); (C.C.)
| | - Lionel Adès
- INSERM UMR-S944, Université de Paris, Institut de la Recherche Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis, 75010 Paris, France; (L.A.); (P.F.)
| | - Pierre Fenaux
- INSERM UMR-S944, Université de Paris, Institut de la Recherche Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis, 75010 Paris, France; (L.A.); (P.F.)
| | - Patricia Krief
- INSERM UMR-S1131, Université de Paris, Institut de la Recherche Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis Hôpital, 75010 Paris, France; (P.G.); (F.G.); (S.G.); (M.C.); (L.G.); (N.T.); (S.B.); (S.P.); (L.G.); (C.L.P.); (M.P.); (P.K.); (C.C.)
| | - Christine Chomienne
- INSERM UMR-S1131, Université de Paris, Institut de la Recherche Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis Hôpital, 75010 Paris, France; (P.G.); (F.G.); (S.G.); (M.C.); (L.G.); (N.T.); (S.B.); (S.P.); (L.G.); (C.L.P.); (M.P.); (P.K.); (C.C.)
| | - Nader Omidvar
- Department of Haematology, Cardiff University School of Medicine, Cardiff CF14 4XN, UK;
| | - Rose Ann Padua
- INSERM UMR-S1131, Université de Paris, Institut de la Recherche Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis Hôpital, 75010 Paris, France; (P.G.); (F.G.); (S.G.); (M.C.); (L.G.); (N.T.); (S.B.); (S.P.); (L.G.); (C.L.P.); (M.P.); (P.K.); (C.C.)
- Correspondence: ; Tel.: +33-1-57-27-90-22; Fax: +33-1-57-27-90-13
| |
Collapse
|
190
|
BH3 Mimetics in Hematologic Malignancies. Int J Mol Sci 2021; 22:ijms221810157. [PMID: 34576319 PMCID: PMC8466478 DOI: 10.3390/ijms221810157] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/15/2021] [Accepted: 09/17/2021] [Indexed: 12/28/2022] Open
Abstract
Hematologic malignancies (HM) comprise diverse cancers of lymphoid and myeloid origin, including lymphomas (approx. 40%), chronic lymphocytic leukemia (CLL, approx. 15%), multiple myeloma (MM, approx. 15%), acute myeloid leukemia (AML, approx. 10%), and many other diseases. Despite considerable improvement in treatment options and survival parameters in the new millennium, many patients with HM still develop chemotherapy-refractory diseases and require re-treatment. Because frontline therapies for the majority of HM (except for CLL) are still largely based on classical cytostatics, the relapses are often associated with defects in DNA damage response (DDR) pathways and anti-apoptotic blocks exemplified, respectively, by mutations or deletion of the TP53 tumor suppressor, and overexpression of anti-apoptotic proteins of the B-cell lymphoma 2 (BCL2) family. BCL2 homology 3 (BH3) mimetics represent a novel class of pro-apoptotic anti-cancer agents with a unique mode of action—direct targeting of mitochondria independently of TP53 gene aberrations. Consequently, BH3 mimetics can effectively eliminate even non-dividing malignant cells with adverse molecular cytogenetic alterations. Venetoclax, the nanomolar inhibitor of BCL2 anti-apoptotic protein has been approved for the therapy of CLL and AML. Numerous venetoclax-based combinatorial treatment regimens, next-generation BCL2 inhibitors, and myeloid cell leukemia 1 (MCL1) protein inhibitors, which are another class of BH3 mimetics with promising preclinical results, are currently being tested in several clinical trials in patients with diverse HM. These pivotal trials will soon answer critical questions and concerns about these innovative agents regarding not only their anti-tumor efficacy but also potential side effects, recommended dosages, and the optimal length of therapy as well as identification of reliable biomarkers of sensitivity or resistance. Effective harnessing of the full therapeutic potential of BH3 mimetics is a critical mission as it may directly translate into better management of the aggressive forms of HM and could lead to significantly improved survival parameters and quality of life in patients with urgent medical needs.
Collapse
|
191
|
Burster T, Traut R, Yermekkyzy Z, Mayer K, Westhoff MA, Bischof J, Knippschild U. Critical View of Novel Treatment Strategies for Glioblastoma: Failure and Success of Resistance Mechanisms by Glioblastoma Cells. Front Cell Dev Biol 2021; 9:695325. [PMID: 34485282 PMCID: PMC8415230 DOI: 10.3389/fcell.2021.695325] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/29/2021] [Indexed: 12/28/2022] Open
Abstract
According to the invasive nature of glioblastoma, which is the most common form of malignant brain tumor, the standard care by surgery, chemo- and radiotherapy is particularly challenging. The presence of glioblastoma stem cells (GSCs) and the surrounding tumor microenvironment protects glioblastoma from recognition by the immune system. Conventional therapy concepts have failed to completely remove glioblastoma cells, which is one major drawback in clinical management of the disease. The use of small molecule inhibitors, immunomodulators, immunotherapy, including peptide and mRNA vaccines, and virotherapy came into focus for the treatment of glioblastoma. Although novel strategies underline the benefit for anti-tumor effectiveness, serious challenges need to be overcome to successfully manage tumorigenesis, indicating the significance of developing new strategies. Therefore, we provide insights into the application of different medications in combination to boost the host immune system to interfere with immune evasion of glioblastoma cells which are promising prerequisites for therapeutic approaches to treat glioblastoma patients.
Collapse
Affiliation(s)
- Timo Burster
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Rebecca Traut
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital, Ulm, Germany
| | - Zhanerke Yermekkyzy
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Katja Mayer
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital, Ulm, Germany
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Joachim Bischof
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital, Ulm, Germany
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital, Ulm, Germany
| |
Collapse
|
192
|
Schneller A, Zojer N, Bolomsky A, Ludwig H. Synergistic interaction between HDAC and MCL-1 inhibitors through downregulation of BCL-XL in multiple myeloma. Haematologica 2021; 106:2516-2521. [PMID: 33910332 PMCID: PMC8409038 DOI: 10.3324/haematol.2020.277152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Indexed: 11/09/2022] Open
Affiliation(s)
- Anja Schneller
- Department of Medicine I, Wilhelminen Cancer Research Institute, Clinic Ottakring, Vienna, Austria; Recipient of a DOC Fellowship of the Austrian Academy of Sciences at the Wilhelminen Cancer Research Institute
| | - Niklas Zojer
- Department of Medicine I, Wilhelminen Cancer Research Institute, Clinic Ottakring, Vienna
| | - Arnold Bolomsky
- Department of Medicine I, Wilhelminen Cancer Research Institute, Clinic Ottakring, Vienna
| | - Heinz Ludwig
- Department of Medicine I, Wilhelminen Cancer Research Institute, Clinic Ottakring, Vienna.
| |
Collapse
|
193
|
Photodynamic Therapy Combined with Bcl-2/Bcl-xL Inhibition Increases the Noxa/Mcl-1 Ratio Independent of Usp9X and Synergistically Enhances Apoptosis in Glioblastoma. Cancers (Basel) 2021; 13:cancers13164123. [PMID: 34439278 PMCID: PMC8393699 DOI: 10.3390/cancers13164123] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 07/29/2021] [Indexed: 01/14/2023] Open
Abstract
Simple Summary Glioblastoma represents one of the most common malignant brain tumors in adults and is associated with a poor clinical outcome despite current therapeutic measures. Therefore, novel strategies for the treatment of this disease are urgently needed. In this work, we examined the antineoplastic effects of a combined treatment with photodynamic therapy and ABT-263 on different glioblastoma cells. Photodynamic therapy uses the selective uptake of a photosensitive molecule followed by activation by light of a specific wavelength to kill cancer cells. ABT-263 is a small molecule inhibitor that targets cancer cells by facilitating programmed cell death. This novel combinatorial therapeutic strategy synergistically killed glioblastoma cells. These results indicate that a combination of the two treatment modalities may be of benefit for the treatment of glioblastoma supporting further studies. Abstract The purpose of this study was to assess in vitro whether the biological effects of 5-aminolevulinic acid (5-ALA)-based photodynamic therapy are enhanced by inhibition of the anti-apoptotic Bcl-2 family proteins Bcl-2 and Bcl-xL in different glioblastoma models. Pre-clinical testing of a microcontroller-based device emitting light of 405 nm wavelength in combination with exposure to 5-ALA (PDT) and the Bcl-2/Bcl-xL inhibitor ABT-263 (navitoclax) was performed in human established and primary cultured glioblastoma cells as well as glioma stem-like cells. We applied cell count analyses to assess cellular proliferation and Annexin V/PI staining to examine pro-apoptotic effects. Western blot analyses and specific knockdown experiments using siRNA were used to examine molecular mechanisms of action. Bcl-2/Bcl-xL inhibition synergistically enhanced apoptosis in combination with PDT. This effect was caspase-dependent. On the molecular level, PDT caused an increased Noxa/Mcl-1 ratio, which was even more pronounced when combined with ABT-263 in a Usp9X-independent manner. Our data showed that Bcl-2/Bcl-xL inhibition increases the response of glioblastoma cells toward photodynamic therapy. This effect can be partly attributed to cytotoxicity and is likely related to a pro-apoptotic shift because of an increased Noxa/Mcl-1 ratio. The results of this study warrant further investigation.
Collapse
|
194
|
Daguer JP, Gonse A, Shchukin Y, Farrera-Soler L, Barluenga S, Winssinger N. Dual Bcl-X L /Bcl-2 inhibitors discovered from DNA-encoded libraries using a fragment pairing strategy. Bioorg Med Chem 2021; 44:116282. [PMID: 34216984 DOI: 10.1016/j.bmc.2021.116282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/09/2021] [Accepted: 06/16/2021] [Indexed: 11/26/2022]
Abstract
A dual Bcl-XL / Bcl-2 inhibitor was discovered from DNA-encoded libraries using a two steps process. In the first step, DNA was used to pair PNA-encoded fragments exploring > 250 000 combinations. In the second step, a focused library combining the selected fragments with linkers of different lengths and geometries led to the identification of tight binding adducts that were further investigated for their selective target engagement in pull-down assays, for their affinity by SPR, and their selectivity in a cytotoxicity assay. The best compound showed comparable cellular activity to venetoclax, the first-in-class therapeutic targeting Bcl-2.
Collapse
Affiliation(s)
- Jean-Pierre Daguer
- Department of Organic Chemistry, Faculty of Sciences, NCCR Chemical Biology, University of Geneva, 1211 Geneva, Switzerland
| | - Arthur Gonse
- Department of Organic Chemistry, Faculty of Sciences, NCCR Chemical Biology, University of Geneva, 1211 Geneva, Switzerland
| | - Yevhenii Shchukin
- Department of Organic Chemistry, Faculty of Sciences, NCCR Chemical Biology, University of Geneva, 1211 Geneva, Switzerland
| | - Lluc Farrera-Soler
- Department of Organic Chemistry, Faculty of Sciences, NCCR Chemical Biology, University of Geneva, 1211 Geneva, Switzerland
| | - Sofia Barluenga
- Department of Organic Chemistry, Faculty of Sciences, NCCR Chemical Biology, University of Geneva, 1211 Geneva, Switzerland
| | - Nicolas Winssinger
- Department of Organic Chemistry, Faculty of Sciences, NCCR Chemical Biology, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
195
|
BCL2 inhibitors and MCL1 inhibitors for hematological malignancies. Blood 2021; 138:1120-1136. [PMID: 34320168 DOI: 10.1182/blood.2020006785] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/17/2021] [Indexed: 11/20/2022] Open
Abstract
BCL2 and MCL1 are commonly expressed pro-survival (anti-apoptotic) proteins in hematological cancers and play important roles in their biology either through dysregulation or by virtue of intrinsic importance to the cell-of-origin of the malignancy. A new class of small molecule anti-cancer drugs, BH3-mimetics, now enable specific targeting of these proteins in patients. BH3-mimetics act by inhibiting the pro-survival BCL2 proteins to enable the activation of BAX and BAK, apoptosis effectors which permeabilize the outer mitochondrial membrane, triggering apoptosis directly in many cells and sensitizing others to cell death when combined with other anti-neoplastic drugs. Venetoclax, a specific inhibitor of BCL2, is the first approved in class, demonstrating striking single agent activity in chronic lymphocytic leukemia (CLL) and in other lymphoid neoplasms, as well as activity against acute myeloid leukemia (AML), especially when used in combination. Key insights from the venetoclax experience include that responses occur rapidly, with major activity as monotherapy proving to be the best indicator for success in combination regimens. This emphasizes the importance of adequate single agent studies for drugs in this class. Furthermore, secondary resistance is common with long-term exposure and often mediated by genetic or adaptive changes in the apoptotic pathway, suggesting that BH3-mimetics are better suited to limited-duration, rather than continuous, therapy. The success of venetoclax has inspired development of BH3-mimetics targeting MCL1. Despite promising preclinical activity against MYC-driven lymphomas, myeloma and AML, their success may particularly depend on their tolerability profile given physiological roles for MCL1 in several non-hematological tissues.
Collapse
|
196
|
Senescence and senolytics in cardiovascular disease: Promise and potential pitfalls. Mech Ageing Dev 2021; 198:111540. [PMID: 34237321 PMCID: PMC8387860 DOI: 10.1016/j.mad.2021.111540] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/28/2021] [Accepted: 07/04/2021] [Indexed: 02/08/2023]
Abstract
Ageing is the biggest risk factor for impaired cardiovascular health, with cardiovascular disease being the cause of death in 40 % of individuals over 65 years old. Ageing is associated with an increased prevalence of atherosclerosis, coronary artery stenosis and subsequent myocardial infarction, thoracic aortic aneurysm, valvular heart disease and heart failure. An accumulation of senescence and increased inflammation, caused by the senescence-associated secretory phenotype, have been implicated in the aetiology and progression of these age-associated diseases. Recently it has been demonstrated that compounds targeting components of anti-apoptotic pathways expressed by senescent cells can preferentially induce senescence cells to apoptosis and have been termed senolytics. In this review, we discuss the evidence demonstrating that senescence contributes to cardiovascular disease, with a particular focus on studies that indicate the promise of senotherapy. Based on these data we suggest novel indications for senolytics as a treatment of cardiovascular diseases which have yet to be studied in the context of senotherapy. Finally, while the potential benefits are encouraging, several complications may result from senolytic treatment. We, therefore, consider these challenges in the context of the cardiovascular system.
Collapse
|
197
|
Cellular Senescence in Lung Fibrosis. Int J Mol Sci 2021; 22:ijms22137012. [PMID: 34209809 PMCID: PMC8267738 DOI: 10.3390/ijms22137012] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/13/2021] [Accepted: 06/24/2021] [Indexed: 12/19/2022] Open
Abstract
Fibrosing interstitial lung diseases (ILDs) are chronic and ultimately fatal age-related lung diseases characterized by the progressive and irreversible accumulation of scar tissue in the lung parenchyma. Over the past years, significant progress has been made in our incomplete understanding of the pathobiology underlying fibrosing ILDs, in particular in relation to diverse age-related processes and cell perturbations that seem to lead to maladaptation to stress and susceptibility to lung fibrosis. Growing evidence suggests that a specific biological phenomenon known as cellular senescence plays an important role in the initiation and progression of pulmonary fibrosis. Cellular senescence is defined as a cell fate decision caused by the accumulation of unrepairable cellular damage and is characterized by an abundant pro-inflammatory and pro-fibrotic secretome. The senescence response has been widely recognized as a beneficial physiological mechanism during development and in tumour suppression. However, recent evidence strengthens the idea that it also drives degenerative processes such as lung fibrosis, most likely by promoting molecular and cellular changes in chronic fibrosing processes. Here, we review how cellular senescence may contribute to lung fibrosis pathobiology, and we highlight current and emerging therapeutic approaches to treat fibrosing ILDs by targeting cellular senescence.
Collapse
|
198
|
Ando H, Murakami Y, Eshima K, Ishida T. A novel polyethylene glycol (PEG)-drug conjugate of Venetoclax, a Bcl-2 inhibitor, for treatment of acute myeloid leukemia (AML). Cancer Rep (Hoboken) 2021; 5:e1485. [PMID: 34173723 PMCID: PMC8955075 DOI: 10.1002/cnr2.1485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 05/16/2021] [Accepted: 06/01/2021] [Indexed: 11/09/2022] Open
Abstract
Background Venetoclax (VTX) is an anticancer drug. It is a selective Bcl‐2 inhibitor that is clinically used for the treatment of patients with lymphomas and leukemias. Treatment with VTX, however, is accompanied by severe adverse events such as tumor lysis syndrome and neutropenia, because VTX readily binds to serum proteins, which results in poor pharmacokinetics and poor tumor tissue concentration. To avoid such adverse events, VTX is administered using a daily or weekly ramp‐up schedule that is cumbersome in clinical situations. Aims To overcome these shortcomings, we prepared a novel polyethylene glycol (PEG)‐drug conjugate of VTX (PEG‐VTX) and evaluated its cytotoxic effects on acute myeloid leukemia (AML) both in vitro and in vivo. Methods and results VTX and 4‐armed PEG derivatives were covalently attached through an amide bond linker. In a series of in vitro studies, PEG‐VTX selectively induced potent growth inhibition of MV4‐11 human AML cells via the inducement of Bcl‐2‐mediated apoptosis. PEG‐VTX had the effect of free VTX, presumably due to the protease‐mediated release of VTX from the conjugates. In in vivo studies with AML tumor‐xenograft mice models, intravenous PEG‐VTX promoted sufficient tumor growth suppression. Compared with a regimen of oral free VTX, the intravenous regimen in those studies used a VTX dosage that was 15–30 times smaller for an OCI‐AML‐2 xenograft model and a dosing regimen that was less frequent for an MV4‐11 xenograft model. The most important development, however, was the absence of weight loss related to severe side effects throughout the treatments. An increase in water solubility and the resultant hydrodynamic size of VTX via PEGylation improved the pharmacokinetics of VTX by avoiding protein interactions and lessening the extravasation from blood. The result was an increase in tumor accumulation and a decrease in the nonspecific distribution of VTX. Conclusion The results of this study suggest that PEG‐VTX could be an alternative therapeutic option for the safe and effective treatment of patients with AML.
Collapse
Affiliation(s)
- Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Yuta Murakami
- Biotechnology & Medical Division, Planning Department, Sanyo Chemical Industries, Ltd, Kyoto, Japan
| | | | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
199
|
Salah HT, DiNardo CD, Konopleva M, Khoury JD. Potential Biomarkers for Treatment Response to the BCL-2 Inhibitor Venetoclax: State of the Art and Future Directions. Cancers (Basel) 2021; 13:2974. [PMID: 34198580 PMCID: PMC8231978 DOI: 10.3390/cancers13122974] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 12/30/2022] Open
Abstract
Intrinsic apoptotic pathway dysregulation plays an essential role in all cancers, particularly hematologic malignancies. This role has led to the development of multiple therapeutic agents targeting this pathway. Venetoclax is a selective BCL-2 inhibitor that has been approved for the treatment of chronic lymphoid leukemia and acute myeloid leukemia. Given the reported resistance to venetoclax, understanding the mechanisms of resistance and the potential biomarkers of response is crucial to ensure optimal drug usage and improved patient outcomes. Mechanisms of resistance to venetoclax include alterations involving the BH3-binding groove, BCL2 gene mutations affecting venetoclax binding, and activation of alternative anti-apoptotic pathways. Moreover, various potential genetic biomarkers of venetoclax resistance have been proposed, including chromosome 17p deletion, trisomy 12, and TP53 loss or mutation. This manuscript provides an overview of biomarkers that could predict treatment response to venetoclax.
Collapse
Affiliation(s)
- Haneen T. Salah
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
| | - Courtney D. DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.D.D.); (M.K.)
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (C.D.D.); (M.K.)
| | - Joseph D. Khoury
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
200
|
Tao ZF, Wang X, Chen J, Ingram JP, Jin S, Judge RA, Kovar PJ, Park C, Sun C, Wakefield BD, Zhou L, Zhang H, Elmore SW, Phillips DC, Judd AS, Leverson JD, Souers AJ. Structure-Based Design of A-1293102, a Potent and Selective BCL-X L Inhibitor. ACS Med Chem Lett 2021; 12:1011-1016. [PMID: 34141086 PMCID: PMC8201748 DOI: 10.1021/acsmedchemlett.1c00162] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/07/2021] [Indexed: 01/10/2023] Open
Abstract
BCL-XL, an antiapoptotic member of the BCL-2 family of proteins, drives tumor survival and maintenance and thus represents a key target for cancer treatment. Herein we report the rational design of a novel series of selective BCL-XL inhibitors exemplified by A-1293102. This molecule contains structural elements of selective BCL-XL inhibitor A-1155463 and the dual BCL-XL/BCL-2 inhibitors ABT-737 and navitoclax, while representing a distinct pharmacophore as assessed by an objective cheminformatic evaluation. A-1293102 exhibited picomolar binding affinity to BCL-XL and both efficiently and selectively killed BCL-XL-dependent tumor cells. X-ray crystallographic analysis demonstrated a key hydrogen bonding network in the P2 binding pocket of BCL-XL, while the bent-back moiety achieved efficient occupancy of the P4 pocket in a manner similar to that of navitoclax. A-1293102 represents one of the few distinct structural series of selective BCL-XL inhibitors, and thus serves as a useful tool for biological studies as well as a lead compound for further optimization.
Collapse
Affiliation(s)
- Zhi-Fu Tao
- AbbVie Inc., 1 North Waukegan Rd, North
Chicago, Illinois 60064, United States
| | - Xilu Wang
- AbbVie Inc., 1 North Waukegan Rd, North
Chicago, Illinois 60064, United States
| | - Jun Chen
- AbbVie Inc., 1 North Waukegan Rd, North
Chicago, Illinois 60064, United States
| | - Justin P. Ingram
- AbbVie Inc., 1 North Waukegan Rd, North
Chicago, Illinois 60064, United States
| | - Sha Jin
- AbbVie Inc., 1 North Waukegan Rd, North
Chicago, Illinois 60064, United States
| | - Russell A. Judge
- AbbVie Inc., 1 North Waukegan Rd, North
Chicago, Illinois 60064, United States
| | - Peter J. Kovar
- AbbVie Inc., 1 North Waukegan Rd, North
Chicago, Illinois 60064, United States
| | - Chang Park
- AbbVie Inc., 1 North Waukegan Rd, North
Chicago, Illinois 60064, United States
| | - Chaohong Sun
- AbbVie Inc., 1 North Waukegan Rd, North
Chicago, Illinois 60064, United States
| | - Brian D. Wakefield
- AbbVie Inc., 1 North Waukegan Rd, North
Chicago, Illinois 60064, United States
| | - Li Zhou
- AbbVie Inc., 1 North Waukegan Rd, North
Chicago, Illinois 60064, United States
| | - Haichao Zhang
- AbbVie Inc., 1 North Waukegan Rd, North
Chicago, Illinois 60064, United States
| | - Steven W. Elmore
- AbbVie Inc., 1 North Waukegan Rd, North
Chicago, Illinois 60064, United States
| | - Darren C. Phillips
- AbbVie Inc., 1 North Waukegan Rd, North
Chicago, Illinois 60064, United States
| | - Andrew S. Judd
- AbbVie Inc., 1 North Waukegan Rd, North
Chicago, Illinois 60064, United States
| | - Joel D. Leverson
- AbbVie Inc., 1 North Waukegan Rd, North
Chicago, Illinois 60064, United States
| | - Andrew J. Souers
- AbbVie Inc., 1 North Waukegan Rd, North
Chicago, Illinois 60064, United States
| |
Collapse
|