1
|
Yao J, Tang S, Shi C, Lin Y, Ge L, Chen Q, Ou B, Liu D, Miao Y, Xie Q, Tang X, Fei J, Yang G, Tian J, Zeng X. Isoginkgetin, a potential CDK6 inhibitor, suppresses SLC2A1/GLUT1 enhancer activity to induce AMPK-ULK1-mediated cytotoxic autophagy in hepatocellular carcinoma. Autophagy 2023; 19:1221-1238. [PMID: 36048765 PMCID: PMC10012924 DOI: 10.1080/15548627.2022.2119353] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/02/2022] Open
Abstract
Isoginkgetin (ISO), a natural biflavonoid, exhibited cytotoxic activity against several types of cancer cells. However, its effects on hepatocellular carcinoma (HCC) cells and mechanism remain unclear. Here, we revealed that ISO effectively inhibited HCC cell proliferation and migration in vitro. LC3-II expression and autophagosomes were increased under ISO treatment. In addition, ISO-induced cell death was attenuated by treatment with chloroquine or knockdown of autophagy-related genes (ATG5 or ULK1). ISO significantly suppressed SLC2A1/GLUT1 (solute carrier family 2 member 1) expression and glucose uptake, leading to activation of the AMPK-ULK1 axis in HepG2 cells. Overexpression of SLC2A1/GLUT1 abrogated ISO-induced autophagy. Combining molecular docking with thermal shift analysis, we confirmed that ISO directly bound to the N terminus of CDK6 (cyclin-dependent kinase 6) and promoted its degradation. Overexpression of CDK6 abrogated ISO-induced inhibition of SLC2A1/GLUT1 transcription and induction of autophagy. Furthermore, ISO treatment significantly decreased the H3K27ac, H4K8ac and H3K4me1 levels on the SLC2A1/GLUT1 enhancer in HepG2 cells. Finally, ISO suppressed the hepatocarcinogenesis in the HepG2 xenograft mice and the diethylnitrosamine+carbon tetrachloride (DEN+CCl4)-induced primary HCC mice and we confirmed SLC2A1/GLUT1 and CDK6 as promising oncogenes in HCC by analysis of TCGA data and human HCC tissues. Our results provide a new molecular mechanism by which ISO treatment or CDK6 deletion promotes autophagy; that is, ISO targeting the N terminus of CDK6 for degradation inhibits the expression of SLC2A1/GLUT1 by decreasing the enhancer activity of SLC2A1/GLUT1, resulting in decreased glucose levels and inducing the AMPK-ULK1 pathway.
Collapse
Affiliation(s)
- Jie Yao
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
- Department of Biochemistry and Molecular Biology, Medical College of Jinan University, Guangzhou, Guangdong, China
| | - Shuming Tang
- Department of Clinical Laboratory, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Chenyan Shi
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Yunzhi Lin
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Lanlan Ge
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
- Department of pathology(Longhua Branch), Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Qinghua Chen
- Department of Pharmacy, Shenzhen Baoan Authentic TCM Therapy Hospital, Shenzhen, Guangdong, China
| | - Baoru Ou
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Dongyu Liu
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Yuyang Miao
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Qiujie Xie
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Xudong Tang
- Key Lab for New Drug Research of TCM and Guangdong Innovative Chinese Medicine and Natural Medicine Engineering Technology Research Center, Research Institute of Tsinghua University, Shenzhen, Guangdong, China
| | - Jia Fei
- Department of Biochemistry and Molecular Biology, Medical College of Jinan University, Guangzhou, Guangdong, China
| | - Guangyi Yang
- Department of Pharmacy, Shenzhen Baoan Authentic TCM Therapy Hospital, Shenzhen, Guangdong, China
| | - Jun Tian
- College of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu, China
| | - Xiaobin Zeng
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
- Department of Clinical Laboratory, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Medicine School of Shenzhen University, Shenzhen, Guangdong, China
| |
Collapse
|
2
|
Buschner G, Feuerecker B, Spinner S, Seidl C, Essler M. Differentiation of acute myeloid leukemia (AML) cells with ATRA reduces 18F-FDG uptake and increases sensitivity towards ABT-737-induced apoptosis. Leuk Lymphoma 2020; 62:630-639. [PMID: 33140666 DOI: 10.1080/10428194.2020.1839648] [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: 10/23/2022]
Abstract
Acute myeloid leukemia (AML) is a malignant disease of the bone marrow, comprising various subtypes. We have investigated seven different AML cell lines that showed different sensitivities toward the inducer of apoptosis ABT-737, with IC50 concentrations ranging from 9.9 nM to 1.8 µM. Besides, the AML cell lines revealed distinct differences in 18F-FDG uptake ranging from 4.1 to 11.0%. Moreover, the Pearson coefficient (0.363) suggests a moderate correlation between 18F-FDG uptake and the IC50 values of ABT-737. Differentiation of the AML cell lines NB-4 and AML-193 with all-trans-retinoic-acid (ATRA) induced a significant increase in sensitivity towards ABT-737 along with a reduced uptake of 18F-FDG. Therefore, 18F-FDG uptake could be predictive on sensitivity to treatment with ABT-737. Furthermore, because differentiation treatment of AML cells using ATRA reduced 18F-FDG uptake and increased sensitivity towards ABT-737, a combined treatment regimen with ATRA and ABT-737 might be a promising therapeutic option in the future.
Collapse
Affiliation(s)
- Gabriel Buschner
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Benedikt Feuerecker
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Sabine Spinner
- Department of Hematology and Oncology, Internal Medicine III, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Christof Seidl
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Markus Essler
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Department of Nuclear Medicine, Universitätsklinikum Bonn, Bonn, Germany
| |
Collapse
|
3
|
Shahar N, Larisch S. Inhibiting the inhibitors: Targeting anti-apoptotic proteins in cancer and therapy resistance. Drug Resist Updat 2020; 52:100712. [DOI: 10.1016/j.drup.2020.100712] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/29/2020] [Accepted: 06/05/2020] [Indexed: 12/14/2022]
|
4
|
Mamriev D, Abbas R, Klingler FM, Kagan J, Kfir N, Donald A, Weidenfeld K, Sheppard DW, Barkan D, Larisch S. A small-molecule ARTS mimetic promotes apoptosis through degradation of both XIAP and Bcl-2. Cell Death Dis 2020; 11:483. [PMID: 32587235 PMCID: PMC7316745 DOI: 10.1038/s41419-020-2670-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 12/13/2022]
Abstract
Many human cancers over-express B cell lymphoma 2 (Bcl-2) or X-linked inhibitor of apoptosis (IAP) proteins to evade cell death. The pro-apoptotic ARTS (Sept4_i2) protein binds directly to both Bcl-2 and XIAP and promotes apoptosis by stimulating their degradation via the ubiquitin-proteasome system (UPS). Here we describe a small molecule, A4, that mimics the function of ARTS. Microscale thermophoresis assays showed that A4 binds XIAP, but not cellular inhibitor of apoptosis protein 1 (cIAP1). A4 binds to a distinct ARTS binding pocket in the XIAP-BIR3 (baculoviral IAP repeat 3) domain. Like ARTS, A4 stimulated poly-ubiquitylation and UPS-mediated degradation of XIAP and Bcl-2, but not cIAP1, resulting in caspase-9 and -3 activation and apoptosis. In addition, over-expression of XIAP rescued HeLa cells from A4-induced apoptosis, consistent with the idea that A4 kills by antagonizing XIAP. On the other hand, treatment with the SMAC-mimetic Birinapant induced secretion of tumour necrosis factor-α (TNFα) and killed ~50% of SKOV-3 cells, and addition of A4 to Birinapant-treated cells significantly reduced secretion of TNFα and blocked Birinapant-induced apoptosis. This suggests that A4 acts by specifically targeting XIAP. The effect of A4 was selective as peripheral blood mononuclear cells and normal human breast epithelial cells were unaffected. Furthermore, proteome analysis revealed that cancer cell lines with high levels of XIAP were particularly sensitive to the killing effect of A4. These results provide proof of concept that the ARTS binding site in XIAP is "druggable". A4 represents a novel class of dual-targeting compounds stimulating apoptosis by UPS-mediated degradation of important anti-apoptotic oncogenes.
Collapse
Affiliation(s)
- Dana Mamriev
- Cell Death and Cancer Research Laboratory, Department of Human Biology and Medical Sciences, University of Haifa, Haifa, 31905, Israel.,The Laboratory of Tumor Dormancy and Metastasis, Department of Human Biology and Medical Sciences, University of Haifa, Haifa, 31905, Israel
| | - Ruqaia Abbas
- Cell Death and Cancer Research Laboratory, Department of Human Biology and Medical Sciences, University of Haifa, Haifa, 31905, Israel
| | | | - Juliana Kagan
- Cell Death and Cancer Research Laboratory, Department of Human Biology and Medical Sciences, University of Haifa, Haifa, 31905, Israel
| | - Nir Kfir
- Cell Death and Cancer Research Laboratory, Department of Human Biology and Medical Sciences, University of Haifa, Haifa, 31905, Israel
| | | | - Keren Weidenfeld
- The Laboratory of Tumor Dormancy and Metastasis, Department of Human Biology and Medical Sciences, University of Haifa, Haifa, 31905, Israel
| | | | - Dalit Barkan
- The Laboratory of Tumor Dormancy and Metastasis, Department of Human Biology and Medical Sciences, University of Haifa, Haifa, 31905, Israel
| | - Sarit Larisch
- Cell Death and Cancer Research Laboratory, Department of Human Biology and Medical Sciences, University of Haifa, Haifa, 31905, Israel.
| |
Collapse
|
5
|
Bad phosphorylation as a target of inhibition in oncology. Cancer Lett 2017; 415:177-186. [PMID: 29175460 DOI: 10.1016/j.canlet.2017.11.017] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/13/2017] [Accepted: 11/17/2017] [Indexed: 12/19/2022]
Abstract
Bcl-2 agonist of cell death (BAD) is a BH3-only member of the Bcl-2 family which possesses important regulatory function in apoptosis. BAD has also been shown to possess many non-apoptotic functions closely linked to cancer including regulation of glycolysis, autophagy, cell cycle progression and immune system development. Interestingly, BAD can be either pro-apoptotic or pro-survival depending on the phosphorylation state of three specific serine residues (human S75, S99 and S118). Expression of BAD and BAD phosphorylation patterns have been shown to influence tumor initiation and progression and play a predictive role in disease prognosis, drug response and chemosensitivity in various cancers. This review aims to summarize the current evidence on the functional role of BAD phosphorylation in human cancer and evaluate the potential utility of modulating BAD phosphorylation in cancer.
Collapse
|
6
|
Ma J, Zhao Z, Wu K, Xu Z, Liu K. MCL-1 is the key target of adjuvant chemotherapy to reverse the cisplatin-resistance in NSCLC. Gene 2016; 587:147-54. [PMID: 27138804 DOI: 10.1016/j.gene.2016.04.054] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 04/27/2016] [Accepted: 04/27/2016] [Indexed: 10/21/2022]
Abstract
Cisplatin is one of the most effective chemotherapeutic agents for the treatment of lung cancer. However, the acquired resistance occurred in cancer cells limits the clinical application of cisplatin. MCL-1, which is an important member in the pro-survival Bcl-2 family, plays a critical role in multidrug resistance (MDR). The aim of the present study is to investigate the value of Pan-Bcl-2 inhibitor as sensitizer for the chemotherapy of cisplatin-resistant non-small cell lung cancer (NSCLC) cells. We found the obatoclax but not the ABT-737 significantly decreased the IC50 (half maximal inhibitory concentration) of cisplatin in cisplatin-resistant NSCLC cells. Furthermore, we demonstrated that the mechanism of obatoclax-promoted cell death induced by cisplatin was dependent on the inhibition of MCL-1, which couldn't be inhibited by ABT-737 but is the target of obatoclax. Moreover, inhibition of MCL-1 recovered the function of NOXA and BAK in cisplatin-resistant NSCLC cells, leading to the promotion of mitochondrial apoptosis induced by cisplatin. Interestingly, our date indicated the obatoclax also reversed the cross-resistance in cisplatin-resistant NSCLC cells. Therefore, we demonstrated that the targeted therapy with MCL-1 inhibitors, such as obatoclax, may represent a novel strategy for cancer therapy.
Collapse
Affiliation(s)
- Jun Ma
- Thoracic Surgery Department, 1st Affiliated Hospital of Sun Yat-Sen Univesity, Guangzhou 510080, China
| | - Zhenxian Zhao
- Pancreato-Biliary Surgery Department, 1st Affiliated Hospital of Sun Yat-Sen Univesity, Guangzhou 510080, China
| | - Kaiming Wu
- Colorectal Surgery Department, 1st Affiliated Hospital of Sun Yat-Sen Univesity, Guangzhou 510080, China
| | - Zhe Xu
- Division of Cardiac Surgery, 1st Affiliated Hospital of Sun Yat-Sen Univesity, Guangzhou 510080, China
| | - Kuanzhi Liu
- Department of Anaesthesiology, 1st Affiliated Hospital of Sun Yat-Sen Univesity, Guangzhou 510080, China.
| |
Collapse
|
7
|
Song T, Chai G, Liu Y, Yu X, Wang Z, Zhang Z. Bcl-2 phosphorylation confers resistance on chronic lymphocytic leukaemia cells to the BH3 mimetics ABT-737, ABT-263 and ABT-199 by impeding direct binding. Br J Pharmacol 2016; 173:471-83. [PMID: 26493374 PMCID: PMC4728412 DOI: 10.1111/bph.13370] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 10/08/2015] [Accepted: 10/14/2015] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND AND PURPOSE Although the ongoing clinical trials of ABT-263 and ABT-199 in chronic lymphocytic leukaemia (CLL) have indicated that BH3 mimetics hold considerable promise, understanding the mechanism of CLL resistance to BH3 mimetics remains a challenge. EXPERIMENTAL APPROACH The LD50 values of ABT-737, ABT-263 and ABT-199 in a number of primary CLL cells from 40 patients, were determined. The levels of Bcl-2 family proteins, including phosphorylated Bcl-2 (pBcl-2) and their interactions were measured by immunoblotting and co-immunoprecipitation. In vitro binding assays were performed by isothermal titration calorimetry and ELISA. BH3 profiling in isolated mitochondria was analysed. KEY RESULTS The ratio of (Mcl-1 + pBcl-2) to Bcl-2 expression provided the most significant predictive marker for the cytotoxic potential of ABT-737, ABT-263 and ABT-199 in the panel of CLL samples. Mechanistically, pBcl-2 inhibited the effects of the ABT compounds on the displacement of Bax and Bim from Bcl-2, thereby suppressing mitochondrial apoptosis. The ABT compounds exhibited 100-300-fold lower binding affinity to the glutamic acid, phosphomimetic, mutant of Bcl-2 (T69E, S70E and S87E; EEE-Bcl-2). BH3 peptides exhibited different rank orders of binding affinities to full-length WT-Bcl-2 and full-length EEE-Bcl-2. CONCLUSIONS AND IMPLICATIONS Our study suggested that a structural alteration in the BH3-binding groove was induced by phosphorylation of Bcl-2. Our data also provided a framework to overcome resistance of CLL cells to the ABT compounds by combining pBcl-2 kinase inhibitors with the ABT compounds.
Collapse
Affiliation(s)
- Ting Song
- State Key Laboratory of Fine Chemicals, School of ChemistryDalian University of TechnologyDalianChina
| | - Gaobo Chai
- School of Life Science and TechnologyDalian University of TechnologyDalianChina
| | - Yubo Liu
- School of Life Science and TechnologyDalian University of TechnologyDalianChina
| | - Xiaoyan Yu
- School of Life Science and TechnologyDalian University of TechnologyDalianChina
| | - Ziqian Wang
- State Key Laboratory of Fine Chemicals, School of ChemistryDalian University of TechnologyDalianChina
| | - Zhichao Zhang
- State Key Laboratory of Fine Chemicals, School of ChemistryDalian University of TechnologyDalianChina
| |
Collapse
|
8
|
Besbes S, Mirshahi M, Pocard M, Billard C. Strategies targeting apoptosis proteins to improve therapy of chronic lymphocytic leukemia. Blood Rev 2015; 29:345-350. [PMID: 25887155 DOI: 10.1016/j.blre.2015.03.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 03/02/2015] [Accepted: 03/26/2015] [Indexed: 12/21/2022]
Abstract
A typical feature of chronic lymphocytic leukemia (CLL) is the impaired ability of the leukemic cells to execute their apoptotic suicide program. Various strategies have been developed to restore apoptosis in CLL cells ex vivo. This article reviews the strategies targeting proteins that directly regulate the mitochondrial pathway of apoptosis and caspase activation: (i) inhibiting the expression or activity of prosurvival proteins of the Bcl-2 and IAP (inhibitor of apoptosis protein) families, which are overexpressed in CLL cells and (ii) upregulating proapoptotic BH3-only members of the Bcl-2 family (which are antagonists of the prosurvival members). Preclinical and clinical data have revealed that inhibiting the activity of prosurvival Bcl-2 proteins with BH3 mimetics (so-called because they mimic BH3-only proteins) is an attractive strategy for CLL therapy. Recent results suggest that the development of BH3 mimetics capable of directly activating the apoptosis effectors Bax and Bak may also be envisaged.
Collapse
Affiliation(s)
- Samaher Besbes
- INSERM U965, Hôpital Lariboisière, Paris, France; Université Paris Diderot-Paris 7, UMR S965, Paris, France
| | - Massoud Mirshahi
- INSERM U965, Hôpital Lariboisière, Paris, France; Université Paris Diderot-Paris 7, UMR S965, Paris, France
| | - Marc Pocard
- INSERM U965, Hôpital Lariboisière, Paris, France; Université Paris Diderot-Paris 7, UMR S965, Paris, France
| | - Christian Billard
- INSERM U965, Hôpital Lariboisière, Paris, France; Université Paris Diderot-Paris 7, UMR S965, Paris, France.
| |
Collapse
|
9
|
Zhao LL, Liu YF, Peng LJ, Fei AM, Cui W, Miao SC, Hermine O, Gressin R, Khochbin S, Chen SJ, Wang J, Mi JQ. Arsenic trioxide rewires mantle cell lymphoma response to bortezomib. Cancer Med 2015; 4:1754-66. [PMID: 26310857 PMCID: PMC4674002 DOI: 10.1002/cam4.511] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 07/12/2015] [Indexed: 01/20/2023] Open
Abstract
Although most of the mantle cell lymphoma (MCL) patients initially responded well to bortezomib (BTZ), the dose-dependent toxicities have greatly limited the application of BTZ to MCL. To investigate the efficacy and mechanism of arsenic trioxide (ATO) with BTZ in inducing apoptosis of MCL cells, two MCL cell lines, along with primary cells from MCL patients (n = 4), were used. Additionally, the NOD-SCID mice xenograft model of Jeko-1 cells was established to study the anti-MCL mechanisms in an in vivo setting. ATO treatment highly improved BTZ capacity to inhibit proliferation and induce apoptosis of MCL cells. Furthermore, the interaction of Noxa and Mcl-1 leads Bak to release from Mcl-1 or from Bcl-xl, which could further activate Bak and Bax and then induce cell apoptosis. We also found that when lower doses of BTZ were used in combination with ATO, more effective proapoptotic effects in both the cell lines and the primary cells were obtained compared to the effects of BTZ used alone at higher doses. Simultaneously, the combination of these two drugs delayed the tumor growth in mice more effectively than BTZ alone. The cooperative anti-MCL effects of this combination therapy both in vitro and in vivo strongly provided a new strategy to the clinical treatment of MCL.
Collapse
Affiliation(s)
- Ling-Ling Zhao
- State Key Laboratory for Medical Genomics and Department of Hematology, Shanghai Institute of Hematology, Collaborative Innovation Center of Systems Biomedicine, Pôle Sino-Français des Sciences du Vivant et Genomique, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Clinical Laboratory, Shanghai Xuhui Central Hospital, Shanghai, China
| | - Yuan-Fang Liu
- State Key Laboratory for Medical Genomics and Department of Hematology, Shanghai Institute of Hematology, Collaborative Innovation Center of Systems Biomedicine, Pôle Sino-Français des Sciences du Vivant et Genomique, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li-Jun Peng
- State Key Laboratory for Medical Genomics and Department of Hematology, Shanghai Institute of Hematology, Collaborative Innovation Center of Systems Biomedicine, Pôle Sino-Français des Sciences du Vivant et Genomique, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ai-Mei Fei
- State Key Laboratory for Medical Genomics and Department of Hematology, Shanghai Institute of Hematology, Collaborative Innovation Center of Systems Biomedicine, Pôle Sino-Français des Sciences du Vivant et Genomique, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Cui
- State Key Laboratory for Medical Genomics and Department of Hematology, Shanghai Institute of Hematology, Collaborative Innovation Center of Systems Biomedicine, Pôle Sino-Français des Sciences du Vivant et Genomique, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sheng-Chao Miao
- State Key Laboratory for Medical Genomics and Department of Hematology, Shanghai Institute of Hematology, Collaborative Innovation Center of Systems Biomedicine, Pôle Sino-Français des Sciences du Vivant et Genomique, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Olivier Hermine
- Service d'Hématologie Adultes, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Université Paris Descartes, Paris, France
| | - Remy Gressin
- Département d'Onco-Hématologie, Hôpital A Michallon, CHU de Grenoble, Grenoble, France
| | - Saadi Khochbin
- INSERM U823, Institut Albert Bonniot, Faculté de Médecine, Université Grenoble Alpes, La Tronche, France
| | - Sai-Juan Chen
- State Key Laboratory for Medical Genomics and Department of Hematology, Shanghai Institute of Hematology, Collaborative Innovation Center of Systems Biomedicine, Pôle Sino-Français des Sciences du Vivant et Genomique, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Wang
- State Key Laboratory for Medical Genomics and Department of Hematology, Shanghai Institute of Hematology, Collaborative Innovation Center of Systems Biomedicine, Pôle Sino-Français des Sciences du Vivant et Genomique, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Qing Mi
- State Key Laboratory for Medical Genomics and Department of Hematology, Shanghai Institute of Hematology, Collaborative Innovation Center of Systems Biomedicine, Pôle Sino-Français des Sciences du Vivant et Genomique, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Blockade of BCL-2 proteins efficiently induces apoptosis in progenitor cells of high-risk myelodysplastic syndromes patients. Leukemia 2015; 30:112-23. [PMID: 26153654 DOI: 10.1038/leu.2015.179] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 06/24/2015] [Accepted: 06/25/2015] [Indexed: 12/25/2022]
Abstract
Deregulated apoptosis is an identifying feature of myelodysplastic syndromes (MDS). Whereas apoptosis is increased in the bone marrow (BM) of low-risk MDS patients, progression to high-risk MDS correlates with an acquired resistance to apoptosis and an aberrant expression of BCL-2 proteins. To overcome the acquired apoptotic resistance in high-risk MDS, we investigated the induction of apoptosis by inhibition of pro-survival BCL-2 proteins using the BCL-2/-XL/-W inhibitor ABT-737 or the BCL-2-selective inhibitor ABT-199. We characterized a cohort of 124 primary human BM samples from MDS/secondary acute myeloid leukemia (sAML) patients and 57 healthy, age-matched controls. Inhibition of anti-apoptotic BCL-2 proteins was specifically toxic for BM cells from high-risk MDS and sAML patients, whereas low-risk MDS or healthy controls remained unaffected. Notably, ABT-737 or ABT-199 treatment was capable of targeting the MDS stem/progenitor compartment in high-risk MDS/sAML samples as shown by the reduction in CD34(+) cells and the decreased colony-forming capacity. Elevated expression of MCL-1 conveyed resistance against both compounds. Protection by stromal cells only partially inhibited induction of apoptosis. Collectively, our data show that the apoptotic resistance observed in high-risk MDS/sAML cells can be overcome by the ABT-737 or ABT-199 treatment and implies that BH3 mimetics might delay disease progression in higher-risk MDS or sAML patients.
Collapse
|
11
|
Besbes S, Mirshahi M, Pocard M, Billard C. New dimension in therapeutic targeting of BCL-2 family proteins. Oncotarget 2015; 6:12862-12871. [PMID: 25970783 PMCID: PMC4536985 DOI: 10.18632/oncotarget.3868] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 04/01/2015] [Indexed: 01/22/2023] Open
Abstract
Proteins of the BCL-2 family control the mitochondrial pathway of apoptosis. Targeting these proteins proves to be an attractive strategy for anticancer therapy. The biological context is based on the fact that BH3-only members of the family are specific antagonists of prosurvival members. This prompted the identification of "BH3 mimetic" compounds. These small peptides or organic molecules indeed mimic the BH3 domain of BH3-only proteins: by selectively binding and antagonizing prosurvival proteins, they can induce apoptosis in malignant cells. Some small-molecule inhibitors of prosurvival proteins have already entered clinical trials in cancer patients and two of them have shown significant therapeutic effects. The latest developments in the field of targeting BCL-2 family proteins highlight several new antagonists of prosurvival proteins as well as direct activators of proapoptotic proteins. These compounds open up novel prospects for the development of BH3 mimetic anticancer drugs.
Collapse
Affiliation(s)
- Samaher Besbes
- INSERM U 965, Hôpital Lariboisière, Paris, France
- Université Paris Diderot, UMR S965, Paris, France
| | - Massoud Mirshahi
- INSERM U 965, Hôpital Lariboisière, Paris, France
- Université Paris Diderot, UMR S965, Paris, France
| | - Marc Pocard
- INSERM U 965, Hôpital Lariboisière, Paris, France
- Université Paris Diderot, UMR S965, Paris, France
| | - Christian Billard
- INSERM U 965, Hôpital Lariboisière, Paris, France
- Université Paris Diderot, UMR S965, Paris, France
| |
Collapse
|
12
|
Alford SE, Kothari A, Loeff FC, Eichhorn JM, Sakurikar N, Goselink HM, Saylors RL, Jedema I, Falkenburg JHF, Chambers TC. BH3 Inhibitor Sensitivity and Bcl-2 Dependence in Primary Acute Lymphoblastic Leukemia Cells. Cancer Res 2015; 75:1366-75. [PMID: 25649768 DOI: 10.1158/0008-5472.can-14-1849] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 12/29/2014] [Indexed: 12/25/2022]
Abstract
BH3 mimetic drugs may be useful to treat acute lymphoblastic leukemia (ALL) but the sensitivity of primary tumor cells has not been fully evaluated. Here, B-lineage ALL cell cultures derived from a set of primary tumors were studied with respect to sensitivity to the BH3 mimetics ABT-263 and ABT-199 and to Bcl-2 dependence and function. These ALL cells each expressed high levels of Bcl-2 and exhibited great sensitivity to ABT-263 and ABT-199, which induced rapid apoptotic cell death. BH3 profiling indicated that the ALL cultures were Bcl-2 dependent. Coimmunoprecipitation studies revealed a multifaceted role for Bcl-2 in binding proapoptotic partners including Bax, Bak, Bik, and Bim. ABT-263 disrupted Bcl-2:Bim interaction in cells. Mcl-1 overexpression rendered ALL cells resistant to ABT-263 and ABT-199, with Mcl-1 assuming the role of Bcl-2 in binding Bim. Freshly isolated pediatric ALL blasts also expressed high levels of Bcl-2 and exhibited high sensitivity to Bcl-2 inhibition by the BH3 mimetic compounds. Overall, our results showed that primary ALL cultures were both more sensitive to BH3 mimetics and more uniform in their response than established ALL cell lines that have been evaluated previously. Furthermore, the primary cell model characterized here offers a powerful system for preclinical testing of novel drugs and drug combinations to treat ALL.
Collapse
Affiliation(s)
- Sarah E Alford
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Anisha Kothari
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Floris C Loeff
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| | - Joshua M Eichhorn
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Nandini Sakurikar
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Henriette M Goselink
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| | - Robert L Saylors
- Department of Pediatrics, Section of Pediatric Hematology-Oncology, University of Arkansas for Medical Sciences at Arkansas Children's Hospital, Little Rock, Arkansas
| | - Inge Jedema
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Timothy C Chambers
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas.
| |
Collapse
|
13
|
Abstract
Chronic lymphocytic leukemia (CLL) is characterized by a typical defect in apoptosis and is still an incurable disease. Numerous apoptosis inducers have been described. These synthetic compounds and natural products (mainly derived from plants) display antileukemic properties in vitro and in vivo and some have even been tested in the clinic in CLL. They act through several different mechanisms. Most of them involve proteins of the Bcl-2 family, which are the key regulators in triggering the mitochondrial pathway of caspase-dependent apoptosis. Thus, the Mcl-1/Noxa axis appeared as a target. Here I overview natural and synthetic apoptosis inducers and their mechanisms of action in CLL cells. Opportunities for developing novel, apoptosis-based therapeutics are presented.
Collapse
Affiliation(s)
- Christian Billard
- INSERM U 872, Centre de Recherche des Cordeliers, Equipe 18, Paris, France
| |
Collapse
|
14
|
Billard C. Targeting antiapoptotic and proapoptotic proteins for novel chronic lymphocytic leukemia therapeutics. Int J Hematol Oncol 2014. [DOI: 10.2217/ijh.14.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
SUMMARY Deficiency in the apoptotic program is one of the hallmarks of chronic lymphocytic leukemia. Defective apoptosis mainly results from the constitutive activation of survival pathways, which leads to the transcription and overexpression of antiapoptotic factors. The latter include proteins of the Bcl-2 family and members of the IAP family. The strategy of inhibiting the expression or activity of these antiapoptotic factors has been extensively investigated. Conversely, upregulation of proapoptotic proteins, notably BH3-only members of the Bcl-2 family (capable of antagonizing their antiapoptotic counterparts) has also been consistently described. Either mechanism can promote apoptosis in chronic lymphocytic leukemia cells ex vivo. The present article recapitulates the mechanistic data and how they contribute to the development of therapeutic agents targeting apoptosis.
Collapse
Affiliation(s)
- Christian Billard
- INSERM U965, Hôpital Lariboisière, Paris, France
- Université Paris Diderot-Paris 7, UMR S965, Paris, France
| |
Collapse
|
15
|
Mohana-Kumaran N, Hill DS, Allen JD, Haass NK. Targeting the intrinsic apoptosis pathway as a strategy for melanoma therapy. Pigment Cell Melanoma Res 2014; 27:525-39. [PMID: 24655414 DOI: 10.1111/pcmr.12242] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 03/17/2014] [Indexed: 01/02/2023]
Abstract
Melanoma drug resistance is often attributed to abrogation of the intrinsic apoptosis pathway. Targeting regulators of apoptosis is thus considered a promising approach to sensitizing melanomas to treatment. The development of small-molecule inhibitors that mimic natural antagonists of either antiapoptotic members of the BCL-2 family or the inhibitor of apoptosis proteins (IAPs), known as BH3- or SMAC-mimetics, respectively, are helping us to understand the mechanisms behind apoptotic resistance. Studies using BH3-mimetics indicate that the antiapoptotic BCL-2 protein MCL-1 and its antagonist NOXA are particularly important regulators of BCL-2 family signaling, while SMAC-mimetic studies show that both XIAP and the cIAPs must be targeted to effectively induce apoptosis of cancer cells. Although most solid tumors, including melanoma, are insensitive to these mimetic drugs as single agents, combinations with other therapeutics have yielded promising results, and tests combining them with BRAF-inhibitors, which have already revolutionized melanoma treatment, are a clear priority.
Collapse
Affiliation(s)
- Nethia Mohana-Kumaran
- The Centenary Institute, Newtown, NSW, Australia; School of Biological Sciences, Universiti Sains Malaysia, Penang, Malaysia
| | | | | | | |
Collapse
|
16
|
Both leukaemic and normal peripheral B lymphoid cells are highly sensitive to the selective pharmacological inhibition of prosurvival Bcl-2 with ABT-199. Leukemia 2014; 28:1207-15. [PMID: 24402163 DOI: 10.1038/leu.2014.1] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 12/19/2013] [Indexed: 01/14/2023]
Abstract
Overexpression of the prosurvival protein Bcl-2 marks many B-lymphoid malignancies and contributes to resistance to many commonly used chemotherapeutic agents. The first effective BH3 mimetic inhibitors of Bcl-2, ABT-737 and navitoclax, also target Bcl-xL, causing dose-limiting thrombocytopenia. This prompted the development of the Bcl-2-selective antagonist, ABT-199. Here we show that in lymphoid cells, ABT-199 specifically causes Bax/Bak-mediated apoptosis that is triggered principally by the initiator BH3-only protein Bim. As expected, malignant cells isolated from patients with chronic lymphocytic leukaemia are highly sensitive to ABT-199. However, we found that normal, untransformed mature B cells are also highly sensitive to ABT-199, both in vitro and in vivo. By contrast, the B-cell precursors are largely spared, as are cells of myeloid origin. These results pinpoint the probable impact of the pharmacological inhibition of Bcl-2 by ABT-199 on the normal mature haemopoietic cell lineages in patients, and have implications for monitoring during ABT-199 therapy as well as for the clinical utility of this very promising targeted agent.
Collapse
|
17
|
Kelly GL, Grabow S, Glaser SP, Fitzsimmons L, Aubrey BJ, Okamoto T, Valente LJ, Robati M, Tai L, Fairlie WD, Lee EF, Lindstrom MS, Wiman KG, Huang DC, Bouillet P, Rowe M, Rickinson AB, Herold MJ, Strasser A. Targeting of MCL-1 kills MYC-driven mouse and human lymphomas even when they bear mutations in p53. Genes Dev 2014; 28:58-70. [PMID: 24395247 PMCID: PMC3894413 DOI: 10.1101/gad.232009.113] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 11/19/2013] [Indexed: 11/25/2022]
Abstract
The transcriptional regulator c-MYC is abnormally overexpressed in many human cancers. Evasion from apoptosis is critical for cancer development, particularly c-MYC-driven cancers. We explored which anti-apoptotic BCL-2 family member (expressed under endogenous regulation) is essential to sustain c-MYC-driven lymphoma growth to reveal which should be targeted for cancer therapy. Remarkably, inducible Cre-mediated deletion of even a single Mcl-1 allele substantially impaired the growth of c-MYC-driven mouse lymphomas. Mutations in p53 could diminish but not obviate the dependency of c-MYC-driven mouse lymphomas on MCL-1. Importantly, targeting of MCL-1 killed c-MYC-driven human Burkitt lymphoma cells, even those bearing mutations in p53. Given that loss of one allele of Mcl-1 is well tolerated in healthy tissues, our results suggest that therapeutic targeting of MCL-1 would be an attractive therapeutic strategy for MYC-driven cancers.
Collapse
Affiliation(s)
- Gemma L. Kelly
- The Walter and Eliza Hall Institute, Parkville, Victoria 3052, Australia
- School of Cancer Sciences, University of Birmingham College of Medical and Dental Sciences, The University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3050 Australia
| | - Stephanie Grabow
- The Walter and Eliza Hall Institute, Parkville, Victoria 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3050 Australia
| | - Stefan P. Glaser
- The Walter and Eliza Hall Institute, Parkville, Victoria 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3050 Australia
| | - Leah Fitzsimmons
- School of Cancer Sciences, University of Birmingham College of Medical and Dental Sciences, The University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Brandon J. Aubrey
- The Walter and Eliza Hall Institute, Parkville, Victoria 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3050 Australia
| | - Toru Okamoto
- The Walter and Eliza Hall Institute, Parkville, Victoria 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3050 Australia
| | - Liz J. Valente
- The Walter and Eliza Hall Institute, Parkville, Victoria 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3050 Australia
| | - Mikara Robati
- The Walter and Eliza Hall Institute, Parkville, Victoria 3052, Australia
| | - Lin Tai
- The Walter and Eliza Hall Institute, Parkville, Victoria 3052, Australia
| | - W. Douglas Fairlie
- The Walter and Eliza Hall Institute, Parkville, Victoria 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3050 Australia
| | - Erinna F. Lee
- The Walter and Eliza Hall Institute, Parkville, Victoria 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3050 Australia
| | - Mikael S. Lindstrom
- Department of Oncology–Pathology, Karolinska Institute, Cancer Center Karolinska (CCK), SE-171 76 Stockholm, Sweden
| | - Klas G. Wiman
- Department of Oncology–Pathology, Karolinska Institute, Cancer Center Karolinska (CCK), SE-171 76 Stockholm, Sweden
| | - David C.S. Huang
- The Walter and Eliza Hall Institute, Parkville, Victoria 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3050 Australia
| | - Philippe Bouillet
- The Walter and Eliza Hall Institute, Parkville, Victoria 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3050 Australia
| | - Martin Rowe
- School of Cancer Sciences, University of Birmingham College of Medical and Dental Sciences, The University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Alan B. Rickinson
- School of Cancer Sciences, University of Birmingham College of Medical and Dental Sciences, The University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Marco J. Herold
- The Walter and Eliza Hall Institute, Parkville, Victoria 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3050 Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute, Parkville, Victoria 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3050 Australia
| |
Collapse
|
18
|
Hao Y, Zhao S, Wang Z. Targeting the protein-protein interaction between IRS1 and mutant p110α for cancer therapy. Toxicol Pathol 2013; 42:140-7. [PMID: 24178578 DOI: 10.1177/0192623313506794] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Phosphoinositide-3-kinase, catalytic, alpha polypeptide, which encodes the catalytic p110α subunit of phosphatidylinositol 3-kinase α, is the most frequently mutated oncogene in human cancers. Targeting mutant p110α holds great promise for cancer therapy. However, it is challenging to develop p110α isoform-specific inhibitors. Most p110α mutations occur at two hot spot regions: an acidic cluster (E542, E545, and Q546) in the helical domain and a histidine residue (H1047) in the kinase domain. We recently discovered that p110α helical domain mutant proteins, but not the kinase domain mutant proteins, directly associate with insulin receptor substrate 1 (IRS1). Moreover, we demonstrated that disruption of protein-protein interaction between p110α helical domain mutant and IRS1 inhibits the growth of tumors with such mutations. The direct protein interaction between IRS1 and p110α helical domain mutants may provide a more accessible target for developing novel precision cancer therapy.
Collapse
Affiliation(s)
- Yujun Hao
- 1Department of Genetics and Genome Sciences, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | | | | |
Collapse
|
19
|
Caloric restriction modulates Mcl-1 expression and sensitizes lymphomas to BH3 mimetic in mice. Blood 2013; 122:2402-11. [DOI: 10.1182/blood-2013-01-478651] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Key Points
Caloric restriction reduces Mcl-1 expression and sensitizes lymphoma cells to ABT-737 in vivo. Caloric restriction mimetics can sensitize lymphomas to ABT-737–induced death independently of p53 and of the main BH3-only proteins.
Collapse
|
20
|
Abstract
Targeting apoptosis is an attractive approach in cancer therapy. The BH3-only proteins of the BCL-2 family (having only the BCL-2 homology domain BH3) can trigger apoptosis by binding to the prosurvival members of this family and neutralizing their functional activity (sequestration of the proapoptotic Bcl-2 family members). The "BH3 mimetic" concept has prompted the development of small molecules capable of mimicking BH3-only proteins and thus inducing apoptosis. The prototype BH3 mimetic ABT-737 selectively targets the three prosurvival proteins BCL-XL, BCL-2, and BCL-W (but not MCL-1 or A1) and its oral derivative ABT-263 has proved promising in clinical trials. Some putative BH3 mimetics are also tested clinically while others are still being characterized. This article recapitulates the various known BH3 mimetics and presents the recent developments in the field. The latter include (i) the identification of molecular determinants responsible for the specific interactions between BH3 motifs and the binding grooves of prosurvival proteins and (ii) the characterization of new compounds and particularly BH3 mimetics that antagonize either selectively MCL-1 or BCL-2 or a broad range of prosurvival proteins. These data are critical advances toward the discovery of novel anticancer agents.
Collapse
Affiliation(s)
- Christian Billard
- Corresponding Author: Christian Billard, Centre de Recherche des Cordeliers, UMRS 872, Equipe 18, 15 rue de l'Ecole de Médecine, 75006 Paris, France.
| |
Collapse
|
21
|
Abstract
HER2 is a trans-membrane receptor tyrosine kinase that activates multiple growth-promoting signaling pathways including PI3K-AKT and Ras-MAPK. Dysregulation of HER2 is a frequent occurrence in breast cancer that is associated with poor patient outcomes. A primary function of HER2 is suppressing apoptosis to enhance cell survival giving rise to uncontrolled proliferation and tumor growth. There has been much investigation into the mechanisms by which apoptosis is suppressed by HER2 in hopes of finding clinical targets for HER2-positive breast cancers as these cancers often become resistant to therapies that directly target HER2. Several apoptotic mechanisms have been shown to be deregulated in HER2-overexpressing cells with examples in both the intrinsic and extrinsic apoptotic pathways. HER2-mediated activation of PI3K-AKT signaling is required for many of the mechanisms HER2 uses to suppress apoptosis. HER2 overexpression is correlated with increases in anti-apoptotic Bcl-2 proteins including Bcl-2, Bcl-xL, and Mcl-1. HER2 also suppresses p53-mediated apoptosis by upregulation of MDM2 by activation of AKT. In addition, survivin expression is often increased with HER2 overexpression leading to inhibition of caspase activation. There is also recent evidence to suggest HER2 can directly influence apoptosis by translocation to the mitochondria to inhibit cytochrome c release. HER2 can also suppress cellular reaction to death ligands, especially TRAIL-induced apoptosis. Elucidation of the mechanisms of apoptotic suppression by HER2 suggest that clinical treatment will likely need to target multiple components of these pathways as there is redundancy in HER2-mediated cell survival. Several therapies have attempted to target Bcl-2 proteins that have promising pre-clinical results. Next-generation HER2 targeting therapies include irreversible pan-ERBB inhibitors and antibody-drug conjugates, such as T-DM1 that has very promising clinical results thus far. Further investigation should include elucidating mechanisms of resistance to HER2-targeted therapies and targeting of multiple components of HER2-mediated cell survival.
Collapse
Affiliation(s)
- Richard L Carpenter
- Division of Surgical Sciences, Department of Surgery, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | - Hui-Wen Lo
- Division of Surgical Sciences, Department of Surgery, Duke University School of Medicine, Durham, North Carolina 27710, USA; Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina 27710, USA
| |
Collapse
|
22
|
Karlsson H, Karlsson SCH, Lindqvist AC, Fransson M, Paul-Wetterberg G, Nilsson B, Essand M, Nilsson K, Frisk P, Jernberg-Wiklund H, Loskog A, Loskog SIA. Combining CAR T cells and the Bcl-2 family apoptosis inhibitor ABT-737 for treating B-cell malignancy. Cancer Gene Ther 2013; 20:386-93. [PMID: 23788110 DOI: 10.1038/cgt.2013.35] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Accepted: 05/18/2013] [Indexed: 01/04/2023]
Abstract
B-cell malignancies upregulate the B-cell lymphoma 2 (Bcl-2) family inhibitors of the intrinsic apoptosis pathway, making them therapy resistant. However, small-molecule inhibitors of Bcl-2 family members such as ABT-737 restore a functional apoptosis pathway in cancer cells, and its oral analog ABT-263 (Navitoclax) has entered clinical trials. Gene engineered chimeric antigen receptor (CAR) T cells also show promise in B-cell malignancy, and as they induce apoptosis via the extrinsic pathway, we hypothesized that small-molecule inhibitors of the Bcl-2 family may potentiate the efficacy of CAR T cells by engaging both apoptosis pathways. CAR T cells targeting CD19 were generated from healthy donors as well as from pre-B-ALL (precursor-B acute lymphoblastic leukemia) patients and tested together with ABT-737 to evaluate apoptosis induction in five B-cell tumor cell lines. The CAR T cells were effective even if the cell lines exhibited different apoptosis resistance profiles, as shown by analyzing the expression of apoptosis inhibitors by PCR and western blot. When combining T-cell and ABT-737 therapy simultaneously, or with ABT-737 as a presensitizer, tumor cell apoptosis was significantly increased. In conclusion, the apoptosis inducer ABT-737 enhanced the efficacy of CAR T cells and could be an interesting drug candidate to potentiate T-cell therapy.
Collapse
Affiliation(s)
| | - S C H Karlsson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Raychaudhuri S, Das S. Low Probability Activation of Bax/Bak Can Induce Selective Killing of Cancer Cells by Generating Heterogeneity in Apoptosis. JOURNAL OF HEALTHCARE ENGINEERING 2013; 4:47-66. [DOI: 10.1260/2040-2295.4.1.47] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
24
|
Russo M, Spagnuolo C, Volpe S, Tedesco I, Bilotto S, Russo GL. ABT-737 resistance in B-cells isolated from chronic lymphocytic leukemia patients and leukemia cell lines is overcome by the pleiotropic kinase inhibitor quercetin through Mcl-1 down-regulation. Biochem Pharmacol 2013; 85:927-36. [PMID: 23353698 DOI: 10.1016/j.bcp.2013.01.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 01/14/2013] [Accepted: 01/15/2013] [Indexed: 12/22/2022]
Abstract
Chronic lymphocytic leukemia (CLL) is the most frequent form of leukemia in adult population and despite numerous studies, it is considered an incurable disease. Since CLL is characterized by overexpression of pro-survival Bcl-2 family members, treatments with their antagonists, such as ABT-737, represent a promising new therapeutic strategy. ABT-737 is a BH3 mimetic agent which binds Bcl-2, Bcl-XL and Bcl-w with high affinity, while weakly interacts with Mcl-1 and Bfl-1. Previous studies demonstrated that quercetin, a flavonoid naturally present in food and beverages, was able to sensitize B-cells isolated from CLL patients to apoptosis when associated with death ligands or fludarabine, through a mechanism involving Mcl-1 down-regulation. Here, we report that the association between ABT-737 and quercetin synergistically induces apoptosis in B-cells and in five leukemic cell lines (Combination Index <1). Peripheral blood mononuclear cell from healthy donors were not affected by quercetin treatment. The molecular pathways triggered by quercetin have been investigated in HPB-ALL cells, characterized by the highest resistance to both ABT-737 and quercetin when applied as single molecules, but highly sensitivity to the co-treatment. In this cell line, quercetin down-regulated Mcl-1 through the inhibition of PI3K/Akt signaling pathway, leading to Mcl-1 instability. The same mechanism was confirmed in B-cells. These results may open new clinical perspectives based on a translational approach in CLL therapy.
Collapse
Affiliation(s)
- Maria Russo
- Institute of Food Sciences, National Research Council, 83100 Avellino, Italy
| | | | | | | | | | | |
Collapse
|
25
|
Boohaker RJ, Lee MW, Vishnubhotla P, Perez JM, Khaled AR. The use of therapeutic peptides to target and to kill cancer cells. Curr Med Chem 2012; 19:3794-804. [PMID: 22725698 DOI: 10.2174/092986712801661004] [Citation(s) in RCA: 239] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2011] [Revised: 04/27/2012] [Accepted: 05/03/2012] [Indexed: 02/06/2023]
Abstract
Peptide therapeutics is a promising field for emerging anti-cancer agents. Benefits include the ease and rapid synthesis of peptides and capacity for modifications. An existing and vast knowledge base of protein structure and function can be exploited for novel peptide design. Current research focuses on developing peptides that can (1) serve as tumor targeting moieties and (2) permeabilize membranes with cytotoxic consequences. A survey of recent findings reveals significant trends. Amphiphilic peptides with clusters of hydrophobic and cationic residues are features of anti-microbial peptides that confer the ability to eradicate microbes and show considerable anti-cancer toxicity. Peptides that assemble and form pores can disrupt cell or organelle membranes and cause apoptotic or necrotic death. Cell permeable and tumor-homing peptides can carry biologically active cargo to tumors or tumor vasculature. The challenge lies in developing the clinical application of therapeutic peptides. Improving delivery to tumors, minimizing non-specific toxic effects and discerning pharmacokinetic properties are high among the needs to produce a powerful therapeutic peptide for cancer treatment.
Collapse
Affiliation(s)
- R J Boohaker
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd, Orlando, FL 32827, USA
| | | | | | | | | |
Collapse
|
26
|
Zhang S, Li G, Ma X, Wang Y, Liu G, Feng L, Zhao Y, Zhang G, Wu Y, Ye X, Qin B, Lu J. Norcantharidin enhances ABT-737-induced apoptosis in hepatocellular carcinoma cells by transcriptional repression of Mcl-1. Cell Signal 2012; 24:1803-9. [PMID: 22609455 DOI: 10.1016/j.cellsig.2012.05.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Revised: 05/10/2012] [Accepted: 05/11/2012] [Indexed: 01/16/2023]
Abstract
Small-molecule cell-permeable Bcl-2/Bcl-xL antagonist ABT-737 has recently emerged as a novel cancer therapeutic agent because it potently induces apoptosis in certain cancer cells. However, since ABT-737 binds to Mcl-1 with low affinity, ABT-737-mediated apoptosis signaling is inhibited in hepatocellular carcinoma (HCC) cells and other solid cancer cells due to the elevated expression of Mcl-1. Accordingly, strategies that target Mcl-1 are explored for overcoming ABT-737-resistance. In this study, we reported that Norcantharidin (NCTD), a small-molecule anticancer drug derived from Chinese traditional medicine blister beetle (Mylabris), induced transcriptional repression of Mcl-1 and considerably enhanced ABT-737-triggered cell viability inhibition and apoptosis in multiple HCC cell lines. Moreover, we observed that the enhancement of ABT-737-mediated apoptosis by NCTD was associated with activation of mitochondrial apoptosis signaling pathway, which involved cytosolic release of cytochrome c, cleavage of caspase-9 and caspase-3. Additionally, knockdown of Bax/Bak, the key effectors permeabilizing mitochondrial outer membrane significantly attenuated the enhancement, indicating mitochondrial apoptosis pathway played an essential role in the execution of the apoptosis. Finally, knockdown of Mcl-1 substantially potentiated ABT-737-mediated apoptotic cell death, confirming the potency of Mcl-1 repression by NCTD in enhancing ABT-737-induced apoptosis. These results therefore suggest that combination treatment with NCTD can overcome ABT-737 resistance and enhance ABT-737 therapeutic efficacy in treating human HCC.
Collapse
Affiliation(s)
- Shuijun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Arbel N, Ben-Hail D, Shoshan-Barmatz V. Mediation of the antiapoptotic activity of Bcl-xL protein upon interaction with VDAC1 protein. J Biol Chem 2012; 287:23152-61. [PMID: 22589539 DOI: 10.1074/jbc.m112.345918] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The mitochondrial protein, the voltage-dependent anion channel (VDAC), is implicated in the control of apoptosis, including via its interaction with the pro- and antiapoptotic proteins. We previously demonstrated the direct interaction of Bcl2 with VDAC, leading to reduced channel conductance. VDAC1-based peptides interacted with Bcl2 to prevent its antiapoptotic activity. Here, using a variety of approaches, we show the interaction of the antiapoptotic protein, Bcl-xL, with VDAC1 and reveal that this interaction mediates Bcl-xL protection against apoptosis. C-terminally truncated Bcl-xL(Δ21) interacts with purified VDAC1, as revealed by microscale thermophoresis and as reflected in the reduced channel conductivity of bilayer-reconstituted VDAC1. Overexpression of Bcl-xL prevented staurosporine-induced apoptosis in cells expressing native VDAC1 but not certain VDAC1 mutants. Having identified mutations in VDAC1 that interfere with the Bcl-xL interaction, certain peptides representing VDAC1 sequences, including the N-terminal domain, were designed and generated as recombinant and synthetic peptides. The VDAC1 N-terminal region and two internal sequences were found to bind specifically, and in a concentration- and time-dependent manner, to immobilized Bcl-xL(Δ21), as revealed by surface plasmon resonance. Moreover, expression of the recombinant peptides in cells overexpressing Bcl-xL prevented protection offered by the protein against staurosporine-induced apoptosis. These results point to Bcl-xL acting as antiapoptotic protein, promoting tumor cell survival via binding to VDAC1. These findings suggest that interfering with Bcl-xL binding to the mitochondria by VDAC1-based peptides may serve to induce apoptosis in cancer cells and to potentiate the efficacy of conventional chemotherapeutic agents.
Collapse
Affiliation(s)
- Nir Arbel
- Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | | | | |
Collapse
|
28
|
Nash PD. Why modules matter. FEBS Lett 2012; 586:2572-4. [PMID: 22710154 DOI: 10.1016/j.febslet.2012.04.049] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Revised: 04/23/2012] [Accepted: 04/24/2012] [Indexed: 11/29/2022]
Abstract
The serendipitous discovery of the SH2 domain unleashed a sea-change in our conceptual molecular understanding of protein function. The reductionist approaches that followed from the recognition of modular protein interaction domains transformed our understanding of cellular signal transduction systems, how they evolve and how they may be manipulated. We now recognize thousands of conserved protein modules - many of which have been described in structure and function, implicated in disease, or underlie targeted therapeutics. The reductionist study of isolated protein modules has enabled the reconstruction of the protein interaction networks that underlie cellular signalling. Protein modules themselves are becoming tools to probe cellular activation states and identify key interactions hubs in both normal and diseased cells and the concept of protein modularity is central to the field of synthetic biology. This brief word of introduction serves to highlight the historical impact of the very powerful idea of protein modules and sets the stage for the exciting on-going discoveries discussed in this issue.
Collapse
Affiliation(s)
- Piers D Nash
- Ben May Department for Cancer Research and the Committee on Cancer Biology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
29
|
Billard C. Development of Noxa-like BH3 mimetics for apoptosis-based therapeutic strategy in chronic lymphocytic leukemia. Mol Cancer Res 2012; 10:673-6. [PMID: 22466256 DOI: 10.1158/1541-7786.mcr-12-0015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Despite real advances made in chemoimmunotherapy, chronic lymphocytic leukemia (CLL) is still an incurable disease. New therapeutic strategies based on the restoration of the cell death program seemed relevant. Some members of the Bcl-2 family are critical players in the defective apoptotic program in CLL cells and/or targets of apoptosis inducers in vitro. The concept of BH3 mimetics has led to the characterization of small molecules mimicking proapoptotic BH3-only members of the Bcl-2 family by their ability to bind and antagonize the prosurvival members. Some putative or actual BH3 mimetics are already being tested in clinical trials with somewhat promising results. However, none of them has a high enough interaction affinity with Mcl-1, a crucial antiapoptotic factor in CLL. It has been suggested that resistance to BH3 mimetics can be overcome by using inhibitors of Mcl-1 expression. An alternative and more direct strategy is to design mimetics of the Noxa BH3 domain, which is a specific antagonistic Mcl-1 ligand. The development of such Noxa-like BH3 mimetics, capable of directly interacting with Mcl-1 and efficiently neutralizing its antiapoptotic activity, is extremely important to evaluate their impact on the clinical outcome of patients with CLL.
Collapse
Affiliation(s)
- Christian Billard
- Centre de Recherche des Cordeliers, Equipe 18, INSERM U 872; Université Pierre et Marie Curie, UMRS 872, Paris, France.
| |
Collapse
|
30
|
Billard C. Design of novel BH3 mimetics for the treatment of chronic lymphocytic leukemia. Leukemia 2012; 26:2032-8. [PMID: 22453662 DOI: 10.1038/leu.2012.88] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Impaired programmed cell death is an important factor in the pathogenesis of chronic lymphocytic leukemia (CLL) and in the development of resistance to chemoimmunotherapy. Hence, the reactivation of apoptotic processes is likely to be a pertinent strategy for circumventing this resistance. Proteins from the Bcl-2 family are critical elements in defective apoptosis. Some compounds induce the apoptosis of CLL cells ex vivo by downregulation of prosurvival members of this family (for example, Bcl-2 and Mcl-1), whereas others act by upregulation of proapoptotic Bcl-2 homology (BH) 3-only members (for example, Noxa and Bim). The concept of BH3 mimetics was prompted by the fact that BH3-only proteins are specific antagonistic ligands of prosurvival Bcl-2 family members. This led to the design of small molecules capable of inhibiting the activity of prosurvival Bcl-2 proteins and inducing apoptosis in leukemia cells in vitro and antileukemic effects in animal models. Several putative or actual BH3 mimetics are currently being trialed in the clinic. Two novel BH3 mimetics that can specifically bind to and antagonize Mcl-1 (a crucial antiapoptotic factor in CLL) have recently been discovered. The evaluation of this type of compound's clinical impact in CLL can now be considered.
Collapse
Affiliation(s)
- C Billard
- Centre de Recherche des Cordeliers, UMRS 872 (Equipe 18), Paris, France.
| |
Collapse
|
31
|
Abstract
Apoptosis is a natural process where cells that are no longer required can be eliminated in a highly regulated, controlled manner. Apoptosis is important in maintaining the mammalian immune system and plays a significant role in immune response, positive and negative T cell selection, and cytotoxic death of target cells. When the apoptotic pathways are impaired or are not tightly regulated, autoimmune diseases, inflammatory diseases, viral and bacterial infections and cancers ensue. An imbalance in the anti-apoptotic and pro-apoptotic factors has been implicated in these diseases. Moreover, current therapies directed towards these diseases focus on the modulation of the apoptotic death pathways to regulate the immune response. In this review, we will focus on the process of T cell activation and apoptosis in autoimmune reactions, in response to tumor progression as well as in response to bacterial and viral infections.
Collapse
Affiliation(s)
- Anuradha K Murali
- Departments of Surgery, Medical University of South Carolina, Charleston, SC 29425
| | | |
Collapse
|
32
|
|