1
|
Nguyen HDT, LE TM, Lee D, Cho HJ, Lee J, Sohn SK, Kim JG, Ham JY, Moon JH, Han HS, Kim J, Baek DW. Synergistic Effect of Venetoclax and Bendamustine in Early T-cell Precursor Acute Lymphoblastic Leukemia. In Vivo 2024; 38:1740-1749. [PMID: 38936885 PMCID: PMC11215584 DOI: 10.21873/invivo.13624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND/AIM To date, therapeutic options for T-cell acute lymphoblastic leukemia (T-ALL) remain very limited. This study evaluated the efficacy of monotherapies and combination therapies including a selective BCL-2 inhibitor for T-ALL cell lines, namely Jurkat, CCRF-CEM, and Loucy. MATERIALS AND METHODS Loucy is an early T-precursor ALL (ETP-ALL) cell line characterized by an immature phenotype, whereas Jurkat and CCRF-CEM are late T-cell progenitor ALL (LTP-ALL) cell lines. Monotherapy was conducted with venetoclax, cytarabine, bendamustine, or azacytidine, whereas combination therapy was performed with venetoclax plus cytarabine, venetoclax plus bendamustine, or venetoclax plus azacytidine. Cell viability assay was conducted after 48 h using Trypan blue and the 3-(4, 5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS). Statistical analysis for evaluating synergistic interactions between anticancer drugs was performed by using the SynergyFinder Plus and drc R package. RESULTS Adding venetoclax to cytarabine, bendamustine, or azacitidine achieved an additive effect, with Loewe synergic scores ranging from -10 to 10 in Jurkat and CCRF-CEM. Conversely, the combination of venetoclax and cytarabine displayed an additive effect (Loewe synergic score: 8.45 and 5.82 with MTS and Trypan blue assays, respectively), whereas venetoclax plus bendamustine or azacitidine exhibited a synergistic effect (Loewe synergic score >10 with MTS assay) in Loucy. Remarkably, the Bliss/Loewe score revealed that the combination of venetoclax and bendamustine was the most synergistic, yielding a score of 13.832±0.55. CONCLUSION The combination of venetoclax and bendamustine demonstrated the greatest synergistic effect in suppressing ETP-ALL cell proliferation. Further studies are warranted to determine the mechanisms for the synergism between venetoclax and bendamustine in high-risk T-ALL.
Collapse
Affiliation(s)
- Hong Duc Thi Nguyen
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu, Republic of Korea
- BK21 Four Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Tan Minh LE
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu, Republic of Korea
- BK21 Four Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Donghyeon Lee
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu, Republic of Korea
- BK21 Four Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Hee Jeong Cho
- Department of Hematology/Oncology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jungmin Lee
- Department of Hematology/Oncology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Sang Kyun Sohn
- Department of Hematology/Oncology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jong Gwang Kim
- Department of Hematology/Oncology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Ji Yeon Ham
- Department of Laboratory Medicine, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Joon Ho Moon
- Department of Hematology/Oncology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Hyung Soo Han
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu, Republic of Korea
- BK21 Four Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Juhyung Kim
- Department of Hematology/Oncology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea;
| | - Dong Won Baek
- Department of Hematology/Oncology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea;
| |
Collapse
|
2
|
Jung YY, Ahn KS, Shen M. Unveiling autophagy complexity in leukemia: The molecular landscape and possible interactions with apoptosis and ferroptosis. Cancer Lett 2024; 582:216518. [PMID: 38043785 DOI: 10.1016/j.canlet.2023.216518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/05/2023]
Abstract
Autophagy is a self-digestion multistep process in which causes the homeostasis through degradation of macromolecules and damaged organelles. The autophagy-mediated tumor progression regulation has been a critical point in recent years, revealing the function of this process in reduction or acceleration of carcinogenesis. Leukemia is a haematological malignancy in which abnormal expansion of hematopoietic cells occurs. The current and conventional therapies from chemotherapy to cell transplantation have failed to appropriately treat the leukemia patients. Among the mechanisms dysregulated in leukemia, autophagy is a prominent one in which can regulate the hallmarks of this tumor. The protective autophagy inhibits apoptosis and ferroptosis in leukemia, while toxic autophagy accelerates cell death. The proliferation and invasion of tumor cells are tightly regulated by the autophagy. The direction of regulation depends on the function of autophagy that is protective or lethal. The protective autophagy accelerates chemoresistance and radio-resistsance. The non-coding RNAs, histone transferases and other pathways such as PI3K/Akt/mTOR are among the regulators of autophagy in leukemia progression. The pharmacological intervention for the inhibition or induction of autophagy by the compounds including sesamine, tanshinone IIA and other synthetic compounds can chance progression of leukemia.
Collapse
Affiliation(s)
- Young Yun Jung
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| | - Mingzhi Shen
- Department of General Medicine, Hainan Hospital of PLA General Hospital, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Sanya, China.
| |
Collapse
|
3
|
Muthumanickam S, Ramachandran B, Boomi P, Jeyakanthan J, Prabu HG, Jegatheswaran S, Premkumar K. Combination of bendamustine-azacitidine against Syk target of breast cancer: an in silico study. J Biomol Struct Dyn 2023; 41:13950-13962. [PMID: 37098715 DOI: 10.1080/07391102.2023.2203259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/28/2023] [Indexed: 04/27/2023]
Abstract
Breast cancer (BC) is the most serious and second leading cause of death in women worldwide. When breast cancer is diagnosed and treated early, the chance of long-term survival is up to 90%. On the other hand, 90% of BC patient deaths are due to metastasis and a lack of effective early diagnosis. The existing conventional chemotherapy provides negative feedback due to transportation barriers towards the action sites, multidrug resistance, poor bio-availability, non-specific delivery and systemic side effects on the healthy tissue. Syk protein Kinase has been reported in BC, as a tumor modulator, providing a pro-survival signal and also by restricting epithelial-mesenchymal transition, enhancing cell-cell interactions and inhibiting migration. In the present study, we explored the possibility of targeting BC by attenuating Syk protein Kinase. Hence, we have conjugated the hydrophobic Bendamustine (BEN) and hydrophilic Azacitidine (AZA) anticancer drugs to evaluate their efficacy against BC. The native drugs (BEN and AZA) and designed drug-drug conjugate (BEN-AZA) were docked with Syk protein. Then, the docked complex was performed for Binding Free Energy and Molecular Dynamics Simulations. Furthermore, DFT and ADME properties were carried out. The results revealed that the designed drug-drug conjugate has a better docking score, ΔGbind and admirable stability throughout the simulation when compared with native drugs. In DFT and ADME analyses, the designed drug-drug conjugate has shown good stereo electronic features and pharmaceutical relevant parameters than that of native drugs. The overall results suggested that the designed drug-drug conjugate may be a suitable candidate for BC treatment.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | | | - Pandi Boomi
- Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| | | | - Halliah Gurumallesh Prabu
- Department of Industrial Chemistry, School of Chemical Sciences, Alagappa University, Karaikudi, Tamil Nadu, India
| | | | - Kumpati Premkumar
- Department of Biomedical Science, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| |
Collapse
|
4
|
Abstract
Background: Acute lymphoblastic leukemia (ALL) is a malignant disease characterized by an excessive number of immature lymphocytes, including immature precursors of both B- and T cells. ALL affects children more often than adults. Immature lymphocytes lead to arrested differentiation and proliferation of cells. Its conventional treatments involve medication with dexamethasone, vincristine, and other anticancer drugs. Although the current first-line drugs can achieve effective treatment, they still cannot prevent the recurrence of some patients with ALL. Treatments have high risk of recurrence especially after the first remission. Currently, novel therapies to treat ALL are in need. Autophagy and apoptosis play important roles in regulating cancer development. Autophagy involves degradation of proteins and organelles, and apoptosis leads to cell death. These phenomena are crucial in cancer progression. Past studies reported that many potential anticancer agents regulate intracellular signaling pathways. Methods: The authors discuss the recent research findings on the role of autophagy and apoptosis in ALL. Results: The autophagy and apoptosis are widely used in the treatment of ALL. Most studies showed that many agents regulate autophagy and apoptosis in ALL cell models, clinical trials, and ALL animal models. Conclusions: In summary, activating autophagy and apoptosis pathways are the main strategies for ALL treatments. For ALL, combining new drugs with traditional chemotherapy and glucocorticoids treatments can achieve the greatest therapeutic effect by activating autophagy and apoptosis.
Collapse
Affiliation(s)
- Fang-Liang Huang
- Children's Medical Center, Taichung Veterans General Hospital, Taichung, Taiwan, ROC.,Department of Physical Therapy, Hungkuang University, Taichung, Taiwan, ROC.,Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, Taiwan, ROC
| | - Sheng-Jie Yu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
| | - Chia-Ling Li
- Children's Medical Center, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
| |
Collapse
|
5
|
Molina MS, Hoffman EA, Stokes J, Kummet N, Smith KA, Baker F, Zúñiga TM, Simpson RJ, Katsanis E. Regulatory Dendritic Cells Induced by Bendamustine Are Associated With Enhanced Flt3 Expression and Alloreactive T-Cell Death. Front Immunol 2021; 12:699128. [PMID: 34249005 PMCID: PMC8264365 DOI: 10.3389/fimmu.2021.699128] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/14/2021] [Indexed: 12/30/2022] Open
Abstract
The growth factor Flt3 ligand (Flt3L) is central to dendritic cell (DC) homeostasis and development, controlling survival and expansion by binding to Flt3 receptor tyrosine kinase on the surface of DCs. In the context of hematopoietic cell transplantation, Flt3L has been found to suppress graft-versus-host disease (GvHD), specifically via host DCs. We previously reported that the pre-transplant conditioning regimen consisting of bendamustine (BEN) and total body irradiation (TBI) results in significantly reduced GvHD compared to cyclophosphamide (CY)+TBI. Pre-transplant BEN+TBI conditioning was also associated with greater Flt3 expression among host DCs and an accumulation of pre-cDC1s. Here, we demonstrate that exposure to BEN increases Flt3 expression on both murine bone marrow-derived DCs (BMDCs) and human monocyte-derived DCs (moDCs). BEN favors development of murine plasmacytoid DCs, pre-cDC1s, and cDC2s. While humans do not have an identifiable equivalent to murine pre-cDC1s, exposure to BEN resulted in decreased plasmacytoid DCs and increased cDC2s. BEN exposure and heightened Flt3 signaling are associated with a distinct regulatory phenotype, with increased PD-L1 expression and decreased ICOS-L expression. BMDCs exposed to BEN exhibit diminished pro-inflammatory cytokine response to LPS and induce robust proliferation of alloreactive T-cells. These proliferative alloreactive T-cells expressed greater levels of PD-1 and underwent increased programmed cell death as the concentration of BEN exposure increased. Alloreactive CD4+ T-cell death may be attributable to pre-cDC1s and provides a potential mechanism by which BEN+TBI conditioning limits GvHD and yields T-cells tolerant to host antigen.
Collapse
Affiliation(s)
- Megan S Molina
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States.,Department of Immunobiology, University of Arizona, Tucson, AZ, United States
| | - Emely A Hoffman
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States
| | - Jessica Stokes
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States
| | - Nicole Kummet
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States.,Department of Molecular & Cellular Biology, University of Arizona, Tucson, AZ, United States
| | - Kyle A Smith
- Department of Physiology, University of Arizona, Tucson, AZ, United States.,Department of Nutritional Sciences, University of Arizona, Tucson, AZ, United States
| | - Forrest Baker
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States.,Department of Nutritional Sciences, University of Arizona, Tucson, AZ, United States
| | - Tiffany M Zúñiga
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ, United States
| | - Richard J Simpson
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States.,Department of Immunobiology, University of Arizona, Tucson, AZ, United States.,Department of Nutritional Sciences, University of Arizona, Tucson, AZ, United States.,The University of Arizona Cancer Center, Tucson, AZ, United States
| | - Emmanuel Katsanis
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States.,Department of Immunobiology, University of Arizona, Tucson, AZ, United States.,The University of Arizona Cancer Center, Tucson, AZ, United States.,Department of Medicine, University of Arizona, Tucson, AZ, United States.,Department of Pathology, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
6
|
Tian Y, Qi H, Wang G, Li L, Zhou D. Anticancer effect of sodium metavanadate on murine breast cancer both in vitro and in vivo. Biometals 2021; 34:557-571. [PMID: 33689084 DOI: 10.1007/s10534-021-00295-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/23/2021] [Indexed: 11/12/2022]
Abstract
Sodium metavanadate (NaVO3) exhibits important physiological effects including insulin-like, chemoprevention and anticancer activity. However, the effects of NaVO3 on breast cancer and underlying mechanisms are still unclear. In this study, our results revealed that NaVO3 was able to inhibit proliferation of murine breast cancer cells 4T1 with IC50 value of 8.19 μM and 1.92 μM at 24 h and 48 h, respectively. The mechanisms underlying the inhibition activity were that NaVO3 could increase reactive oxygen species (ROS) level in a concentration-dependent way, arrest cells at G2/M phase, diminish the mitochondrial membrane potential (MMP), finally promote the progress of apoptosis. Furthermore, NaVO3 also exhibited a dose-dependent anticancer activity in breast cancer-bearing mice that led to the shrinkage of tumor volume (about 50%), lower microvessel density, less propagating cells and more apoptotic cells in vivo, as compared to the saline group. Therefore, NaVO3 may act as a potential chemotherapeutic agent in breast cancer treatment.
Collapse
Affiliation(s)
- Yu Tian
- Department of Occupational Health and Environmental Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Haihui Qi
- Department of Occupational Health and Environmental Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Gang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Li Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Dinglun Zhou
- Department of Occupational Health and Environmental Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
7
|
Targeting TRPV1-mediated autophagy attenuates nitrogen mustard-induced dermal toxicity. Signal Transduct Target Ther 2021; 6:29. [PMID: 33487631 PMCID: PMC7829253 DOI: 10.1038/s41392-020-00389-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 01/30/2023] Open
Abstract
Nitrogen mustard (NM) causes severe vesicating skin injury, which lacks effective targeted therapies. The major limitation is that the specific mechanism of NM-induced skin injury is not well understood. Recently, autophagy has been found to play important roles in physical and chemical exposure-caused cutaneous injuries. However, whether autophagy contributes to NM-induced dermal toxicity is unclear. Herein, we initially confirmed that NM dose-dependently caused cell death and induced autophagy in keratinocytes. Suppression of autophagy by 3-methyladenine, chloroquine, and bafilomycin A1 or ATG5 siRNA attenuated NM-induced keratinocyte cell death. Furthermore, NM increased transient receptor potential vanilloid 1 (TRPV1) expression, intracellular Ca2+ content, and the activities of Ca2+/calmodulin-dependent kinase kinase β (CaMKKβ), AMP-activated protein kinase (AMPK), unc-51-like kinase 1 (ULK1), and mammalian target of rapamycin (mTOR). NM-induced autophagy in keratinocytes was abolished by treatment with inhibitors of TRPV1 (capsazepine), CaMKKβ (STO-609), AMPK (compound C), and ULK1 (SBI-0206965) as well as TRPV1, CaMKKβ, and AMPK siRNA transfection. In addition, an mTOR inhibitor (rapamycin) had no significant effect on NM-stimulated autophagy or cell death of keratinocytes. Finally, the results of the in vivo experiment in NM-treated skin tissues were consistent with the findings of the in vitro experiment. In conclusion, NM-caused dermal toxicity by overactivating autophagy partially through the activation of TRPV1-Ca2+-CaMKKβ-AMPK-ULK1 signaling pathway. These results suggest that blocking TRPV1-dependent autophagy could be a potential treatment strategy for NM-caused cutaneous injury.
Collapse
|
8
|
Hydroxamic acid hybrids as the potential anticancer agents: An Overview. Eur J Med Chem 2020; 205:112679. [PMID: 32791404 DOI: 10.1016/j.ejmech.2020.112679] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/12/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023]
|