1
|
McCorkle JR, Ahn R, Cao CD, Hill KS, Dietrich CS, Kolesar JM. Antineoplastic Drug Synergy of Artesunate with Navitoclax in Models of High-Grade Serous Ovarian Cancer. Cancers (Basel) 2024; 16:1321. [PMID: 38610999 PMCID: PMC11011058 DOI: 10.3390/cancers16071321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Artesunate belongs to a class of medications derived from the sweet wormwood plant (Artemisia annua) known as artemisinins. Artesunate has traditionally been used as a frontline treatment for severe malaria but has also demonstrated antineoplastic activity against various malignancies, including ovarian cancer. Data suggest that artesunate exacerbates cellular oxidative stress, triggering apoptosis. In the current study, we investigated the ability of navitoclax, an inhibitor of the antiapoptotic Bcl-2 protein family, to enhance artesunate efficacy in ovarian cancer cells. Artesunate and navitoclax both demonstrated antiproliferative effects on 2D and 3D ovarian cancer cell models as single agents. Upon combination of navitoclax with artesunate, antineoplastic drug synergy was also observed in each of the 2D cell lines and ovarian tumor organoid models tested. Further investigation of this drug combination using intraperitoneal CAOV3 xenograft models in BALB/scid mice showed that the artesunate/navitoclax doublet was superior to single-agent artesunate and vehicle control treatment. However, it did not outperform single-agent navitoclax. With optimization, this drug combination could provide a new therapeutic option for ovarian cancer and warrants further preclinical investigation.
Collapse
Affiliation(s)
- J. Robert McCorkle
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (J.R.M.); (K.S.H.); (C.S.D.)
| | - Rebecca Ahn
- University of Kentucky College of Medicine, Lexington, KY 40536, USA;
| | - Connie D. Cao
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Kentucky, Lexington, KY 40536, USA;
| | - Kristen S. Hill
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (J.R.M.); (K.S.H.); (C.S.D.)
| | - Charles S. Dietrich
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (J.R.M.); (K.S.H.); (C.S.D.)
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Kentucky, Lexington, KY 40536, USA;
| | - Jill M. Kolesar
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; (J.R.M.); (K.S.H.); (C.S.D.)
- Department of Pharmacy Practice and Science, University of Kentucky College of Pharmacy, Lexington, KY 40536, USA
- Department of Clinical Research, University of Kentucky Markey Cancer Center, Lexington, KY 40536, USA
| |
Collapse
|
2
|
Kagan AB, Moses BS, Lapidus R, Mott BT, Rai G, Anders NM, Hoag SW, Rudek MA, Civin CI. ART714 is a best-in-class antileukemic 2-carbon-linked dimeric artemisinin derivative. Cancer Chemother Pharmacol 2023; 92:39-50. [PMID: 37249624 DOI: 10.1007/s00280-023-04539-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/23/2023] [Indexed: 05/31/2023]
Abstract
PURPOSE It has become increasingly clear that new multiagent combination regimens are required to improve survival rates in acute myeloid leukemia (AML). We recently reported that ART631, a first-in-class 2-carbon-linked artemisinin-derived dimer (2C-ART), was not only efficacious as a component of a novel three-drug combination regimen to treat AML, but, like other synthetic artemisinin derivatives, demonstrated low clinical toxicity. However, we ultimately found ART631 to have suboptimal solubility and stability properties, thus limiting its potential for clinical development. METHODS We assessed 22 additional 2C-ARTs with documented in vivo antimalarial activity for antileukemic efficacy and physicochemical properties. Our strategy involved culling out 2C-ARTs inferior to ART631 with respect to potency, stability, and solubility in vitro, and then validating in vivo pharmacokinetics, pharmacodynamics, and efficacy of one 2C-ART lead compound. RESULTS Of the 22 2C-ARTs, ART714 was found to have the most optimal in vitro solubility, stability, and antileukemic efficacy, both alone and in combination with the BCL2 inhibitor venetoclax (VEN) and the kinase inhibitor sorafenib (SOR). ART714 was also highly effective in combination with VEN and the FMS-like tyrosine kinase 3 inhibitor gilteritinib (GILT) against MOLM14 AML xenografts. CONCLUSION We identified ART714 as our best-in-class antileukemic 2C-ART, based on in vitro potency and pharmacologic properties. We established its in vivo pharmacokinetics and demonstrated its in vitro cooperativity with VEN and SOR and in vivo activities of combinations of ART714, VEN, and GILT. Additional research is indicated to define the optimal niche for the use of ART714 in treatment of AML.
Collapse
Affiliation(s)
- Amanda B Kagan
- Department of Medicine, Division of Clinical Pharmacology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Oncology, School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, 1650 Orleans Street, CRB1 Room 1M52, Baltimore, MD, 21231-1000, USA
| | - Blake S Moses
- Departments of Pediatrics and Physiology, School of Medicine, Center for Stem Cell Biology and Regenerative Medicine, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, BRB14-021, 655 W Baltimore St, Baltimore, MD, 21201, USA
- Keros Therapeutics, Inc., Lexington, MA, USA
| | - Rena Lapidus
- Department of Medicine, School of Medicine, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Bryan T Mott
- Department of Neurosurgery, Wake Forest Baptist Health, Winston-Salem, NC, USA
| | - Ganesha Rai
- Department of Pre-clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Nicole M Anders
- Department of Oncology, School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, 1650 Orleans Street, CRB1 Room 1M52, Baltimore, MD, 21231-1000, USA
- Takeda Pharmaceutical Company, San Diego, CA, USA
| | - Stephen W Hoag
- School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Michelle A Rudek
- Department of Medicine, Division of Clinical Pharmacology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
- Department of Oncology, School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, 1650 Orleans Street, CRB1 Room 1M52, Baltimore, MD, 21231-1000, USA.
| | - Curt I Civin
- Departments of Pediatrics and Physiology, School of Medicine, Center for Stem Cell Biology and Regenerative Medicine, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, BRB14-021, 655 W Baltimore St, Baltimore, MD, 21201, USA.
| |
Collapse
|
3
|
Kagan AB, Moses BS, Mott BT, Rai G, Anders NM, Rudek MA, Civin CI. A Novel 2-Carbon-Linked Dimeric Artemisinin With Potent Antileukemic Activity and Favorable Pharmacology. Front Oncol 2022; 11:790037. [PMID: 35127495 PMCID: PMC8811960 DOI: 10.3389/fonc.2021.790037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/17/2021] [Indexed: 12/02/2022] Open
Abstract
Acute myeloid leukemia (AML) remains a devastating disease, with low cure rates despite intensive standard chemotherapy regimens. In the past decade, targeted antileukemic drugs have emerged from research efforts. Nevertheless, targeted therapies are often effective for only a subset of patients whose leukemias harbor a distinct mutational or gene expression profile and provide only transient antileukemic responses as monotherapies. We previously presented single agent and combination preclinical data for a novel 3-carbon-linked artemisinin-derived dimer (3C-ART), diphenylphosphate analog 838 (ART838), that indicates a promising approach to treat AML, given its demonstrated synergy with targeted antileukemic drugs and large therapeutic window. We now report new data from our initial evaluation of a structurally distinct class of 2-carbon-linked dimeric artemisinin-derived analogs (2C-ARTs) with prior documented in vivo antimalarial activity. These 2C-ARTs have antileukemic activity at low (nM) concentrations, have similar cooperativity with other antineoplastic drugs and comparable physicochemical properties to ART838, and provide a viable path to clinical development.
Collapse
Affiliation(s)
- Amanda B. Kagan
- Department of Medicine, Division of Clinical Pharmacology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Blake S. Moses
- Center for Stem Cell Biology & Regenerative Medicine, Marlene and Stewart Greenebaum Comprehensive Cancer Center, Departments of Pediatrics and Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Bryan T. Mott
- Department of Neurosurgery, Wake Forest Baptist Health, Winston-Salem, NC, United States
| | - Ganesha Rai
- Department of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| | - Nicole M. Anders
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Michelle A. Rudek
- Department of Medicine, Division of Clinical Pharmacology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Curt I. Civin
- Center for Stem Cell Biology & Regenerative Medicine, Marlene and Stewart Greenebaum Comprehensive Cancer Center, Departments of Pediatrics and Physiology, School of Medicine, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
4
|
Yu R, Jin G, Fujimoto M. Dihydroartemisinin: A Potential Drug for the Treatment of Malignancies and Inflammatory Diseases. Front Oncol 2021; 11:722331. [PMID: 34692496 PMCID: PMC8529146 DOI: 10.3389/fonc.2021.722331] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/10/2021] [Indexed: 12/12/2022] Open
Abstract
Dihydroartemisinin (DHA) has been globally recognized for its efficacy and safety in the clinical treatment of malaria for decades. Recently, it has been found that DHA inhibits malignant tumor growth and regulates immune system function in addition to anti-malaria. In parasites and tumors, DHA causes severe oxidative stress by inducing excessive reactive oxygen species production. DHA also kills tumor cells by inducing programmed cell death, blocking cell cycle and enhancing anti-tumor immunity. In addition, DHA inhibits inflammation by reducing the inflammatory cells infiltration and suppressing the production of pro-inflammatory cytokines. Further, genomics, proteomics, metabolomics and network pharmacology of DHA therapy provide the basis for elucidating the pharmacological effects of DHA. This review provides a summary of the recent research progress of DHA in anti-tumor, inhibition of inflammatory diseases and the relevant pharmacological mechanisms. With further research of DHA, it is likely that DHA will become an alternative therapy in the clinical treatment of malignant tumors and inflammatory diseases.
Collapse
Affiliation(s)
- Ran Yu
- Department of Immunology and Pathogenic Biology, Yanbian University Medical College, Yanji, China
| | - Guihua Jin
- Department of Immunology and Pathogenic Biology, Yanbian University Medical College, Yanji, China
| | - Manabu Fujimoto
- Department of Dermatology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Laboratory of Cutaneous Immunology, Osaka University Immunology Frontier Research Center, Osaka, Japan
| |
Collapse
|
5
|
Nor Hisam NS, Ugusman A, Rajab NF, Ahmad MF, Fenech M, Liew SL, Mohamad Anuar NN. Combination Therapy of Navitoclax with Chemotherapeutic Agents in Solid Tumors and Blood Cancer: A Review of Current Evidence. Pharmaceutics 2021; 13:pharmaceutics13091353. [PMID: 34575429 PMCID: PMC8468743 DOI: 10.3390/pharmaceutics13091353] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/25/2021] [Accepted: 08/25/2021] [Indexed: 01/19/2023] Open
Abstract
Combination therapy emerges as a fundamental scheme in cancer. Many targeted therapeutic agents are developed to be used with chemotherapy or radiation therapy to enhance drug efficacy and reduce toxicity effects. ABT-263, known as navitoclax, mimics the BH3-only proteins of the BCL-2 family and has a high affinity towards pro-survival BCL-2 family proteins (i.e., BCL-XL, BCL-2, BCL-W) to induce cell apoptosis effectively. A single navitoclax action potently ameliorates several tumor progressions, including blood and bone marrow cancer, as well as small cell lung carcinoma. Not only that, but navitoclax alone also therapeutically affects fibrotic disease. Nevertheless, outcomes from the clinical trial of a single navitoclax agent in patients with advanced and relapsed small cell lung cancer demonstrated a limited anti-cancer activity. This brings accumulating evidence of navitoclax to be used concomitantly with other chemotherapeutic agents in several solid and non-solid tumors that are therapeutically benefiting from navitoclax treatment in preclinical studies. Initially, we justify the anti-cancer role of navitoclax in combination therapy. Then, we evaluate the current evidence of navitoclax in combination with the chemotherapeutic agents comprehensively to indicate the primary regulator of this combination strategy in order to produce a therapeutic effect.
Collapse
Affiliation(s)
- Nur Syahidah Nor Hisam
- Programme of Biomedical Science, Centre for Toxicology & Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia; (N.S.N.H.); (S.L.L.)
| | - Azizah Ugusman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Nor Fadilah Rajab
- Center for Healthy Ageing & Wellness, Programme of Biomedical Science, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia; (N.F.R.); (M.F.)
| | - Mohd Faizal Ahmad
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Michael Fenech
- Center for Healthy Ageing & Wellness, Programme of Biomedical Science, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia; (N.F.R.); (M.F.)
- Genome Health Foundation, North Brighton, SA 5048, Australia
| | - Sze Ling Liew
- Programme of Biomedical Science, Centre for Toxicology & Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia; (N.S.N.H.); (S.L.L.)
| | - Nur Najmi Mohamad Anuar
- Programme of Biomedical Science, Centre for Toxicology & Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia; (N.S.N.H.); (S.L.L.)
- Correspondence: ; Tel.: +60-13-3845844
| |
Collapse
|
6
|
Antileukemic efficacy of a potent artemisinin combined with sorafenib and venetoclax. Blood Adv 2021; 5:711-724. [PMID: 33560385 DOI: 10.1182/bloodadvances.2020003429] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/28/2020] [Indexed: 12/21/2022] Open
Abstract
Artemisinins are active against human leukemia cell lines and have low clinical toxicity in worldwide use as antimalarials. Because multiagent combination regimens are necessary to cure fully evolved leukemias, we sought to leverage our previous finding that artemisinin analogs synergize with kinase inhibitors, including sorafenib (SOR), by identifying additional synergistic antileukemic drugs with low toxicity. Screening of a targeted antineoplastic drug library revealed that B-cell lymphoma 2 (BCL2) inhibitors synergize with artemisinins, and validation assays confirmed that the selective BCL2 inhibitor, venetoclax (VEN), synergized with artemisinin analogs to inhibit growth and induce apoptotic cell death of multiple acute leukemia cell lines in vitro. An oral 3-drug "SAV" regimen (SOR plus the potent artemisinin-derived trioxane diphenylphosphate 838 dimeric analog [ART838] plus VEN) killed leukemia cell lines and primary cells in vitro. Leukemia cells cultured in ART838 had decreased induced myeloid leukemia cell differentiation protein (MCL1) levels and increased levels of DNA damage-inducible transcript 3 (DDIT3; GADD153) messenger RNA and its encoded CCATT/enhancer-binding protein homologous protein (CHOP), a key component of the integrated stress response. Thus, synergy of the SAV combination may involve combined targeting of MCL1 and BCL2 via discrete, tolerable mechanisms, and cellular levels of MCL1 and DDIT3/CHOP may serve as biomarkers for action of artemisinins and SAV. Finally, SAV treatment was tolerable and resulted in deep responses with extended survival in 2 acute myeloid leukemia (AML) cell line xenograft models, both harboring a mixed lineage leukemia gene rearrangement and an FMS-like receptor tyrosine kinase-3 internal tandem duplication, and inhibited growth in 2 AML primagraft models.
Collapse
|
7
|
Malignant pleural mesothelioma co-opts BCL-X L and autophagy to escape apoptosis. Cell Death Dis 2021; 12:406. [PMID: 33859162 PMCID: PMC8050302 DOI: 10.1038/s41419-021-03668-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 03/22/2021] [Accepted: 03/22/2021] [Indexed: 12/15/2022]
Abstract
Escape from programmed cell death is a hallmark of cancer. In this study, we investigated the anti-apoptotic mechanisms and explored the therapeutic potential of BCL-2 homology domain-3 (BH3) mimetics in malignant pleural mesothelioma (MPM), a lethal thoracic malignancy with an extreme dearth of treatment options. By implementing integrated analysis of functional genomic data of MPM cells and quantitative proteomics of patients’ tumors, we identified BCL-XL as an anti-apoptotic driver that is overexpressed and confers an oncogenic dependency in MPM. MPM cells harboring genetic alterations that inactivate the NF2/LATS1/2 signaling are associated with increased sensitivity to A-1155463, a BCL-XL-selective BH3 mimetic. Importantly, BCL-XL inhibition elicits protective autophagy, and concomitant blockade of BCL-XL and autophagic machinery with A-1155463 and hydroxychloroquine (HCQ), the US Food and Drug Administration (FDA)-approved autophagy inhibitor, synergistically enhances anti-MPM effects in vitro and in vivo. Together, our work delineates the molecular basis underlying resistance to apoptosis and uncovers an evasive mechanism that limits response to BH3 mimetics in MPM, suggesting a novel strategy to target this aggressive disease.
Collapse
|
8
|
Li Q, Ma Q, Cheng J, Zhou X, Pu W, Zhong X, Guo X. Dihydroartemisinin as a Sensitizing Agent in Cancer Therapies. Onco Targets Ther 2021; 14:2563-2573. [PMID: 33880035 PMCID: PMC8053502 DOI: 10.2147/ott.s297785] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 03/18/2021] [Indexed: 01/03/2023] Open
Abstract
Cancer is one of the major threats to human health. Although humans have struggled with cancer for decades, the efficacy of treatments for most tumors is still very limited. Dihydroartemisinin (DHA) is a derivative of artemisinin, a first-line antimalarial drug originally developed in China. Beyond the anti-malarial effect, DHA has also been reported to show anti-inflammatory, anti-parasitosis, and immune-modulating properties in vitro and in vivo. Furthermore, an increasing number of studies report that DHA possesses anticancer activities on a wide range of cancer types both in vitro and in vivo, as well as enhances the efficacy of chemotherapy, targeted therapy, and even radiotherapy. However, the mechanisms of DHA on different tumors differ in various ways. In this review, we intend to summarize how DHA sensitizes cancer cells to anti-cancer therapies, highlight its molecular mechanisms and pharmacological effects in vitro and in vivo as well as in current clinical trials, and discuss potential issues concerning DHA. Hopefully, more attention will be paid to DHA as a sensitizer for cancer therapy in the future.
Collapse
Affiliation(s)
- Qingrong Li
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Qiang Ma
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Jibing Cheng
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Xi Zhou
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Wenjie Pu
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Xiaowu Zhong
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Xiaolan Guo
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000, People's Republic of China.,Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| |
Collapse
|
9
|
Lu X, Efferth T. Repurposing of artemisinin-type drugs for the treatment of acute leukemia. Semin Cancer Biol 2021; 68:291-312. [DOI: 10.1016/j.semcancer.2020.05.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 05/22/2020] [Accepted: 05/22/2020] [Indexed: 12/19/2022]
|
10
|
Smith KH, Budhraja A, Lynch J, Roberts K, Panetta JC, Connelly JP, Turnis ME, Pruett-Miller SM, Schuetz JD, Mullighan CG, Opferman JT. The Heme-Regulated Inhibitor Pathway Modulates Susceptibility of Poor Prognosis B-Lineage Acute Leukemia to BH3-Mimetics. Mol Cancer Res 2020; 19:636-650. [PMID: 33288732 DOI: 10.1158/1541-7786.mcr-20-0586] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/28/2020] [Accepted: 12/02/2020] [Indexed: 11/16/2022]
Abstract
Antiapoptotic MCL1 is one of the most frequently amplified genes in human cancers and elevated expression confers resistance to many therapeutics including the BH3-mimetic agents ABT-199 and ABT-263. The antimalarial, dihydroartemisinin (DHA) translationally represses MCL-1 and synergizes with BH3-mimetics. To explore how DHA represses MCL-1, a genome-wide CRISPR screen identified that loss of genes in the heme synthesis pathway renders mouse BCR-ABL+ B-ALL cells resistant to DHA-induced death. Mechanistically, DHA disrupts the interaction between heme and the eIF2α kinase heme-regulated inhibitor (HRI) triggering the integrated stress response. Genetic ablation of Eif2ak1, which encodes HRI, blocks MCL-1 repression in response to DHA treatment and represses the synergistic killing of DHA and BH3-mimetics compared with wild-type leukemia. Furthermore, BTdCPU, a small-molecule activator of HRI, similarly triggers MCL-1 repression and synergizes with BH3-mimetics in mouse and human leukemia including both Ph+ and Ph-like B-ALL. Finally, combinatorial treatment of leukemia bearing mice with both BTdCPU and a BH3-mimetic extended survival and repressed MCL-1 in vivo. These findings reveal for the first time that the HRI-dependent cellular heme-sensing pathway can modulate apoptosis in leukemic cells by repressing MCL-1 and increasing their responsiveness to BH3-mimetics. This signaling pathway could represent a generalizable mechanism for repressing MCL-1 expression in malignant cells and sensitizing them to available therapeutics. IMPLICATIONS: The HRI-dependent cellular heme-sensing pathway can modulate apoptotic sensitivity in leukemic cells by repressing antiapoptotic MCL-1 and increasing their responsiveness to BH3-mimetics.
Collapse
Affiliation(s)
- Kaitlyn H Smith
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, Tennessee.,Integrated Program in Biomedical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Amit Budhraja
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - John Lynch
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Kathryn Roberts
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - John C Panetta
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jon P Connelly
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, Tennessee.,Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Meghan E Turnis
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, Tennessee.,Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - John D Schuetz
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Joseph T Opferman
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, Tennessee.
| |
Collapse
|
11
|
Mancuso RI, Foglio MA, Olalla Saad ST. Artemisinin-type drugs for the treatment of hematological malignancies. Cancer Chemother Pharmacol 2020; 87:1-22. [PMID: 33141328 DOI: 10.1007/s00280-020-04170-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/06/2020] [Indexed: 12/19/2022]
Abstract
Qinghaosu, known as artemisinin (ARS), has been for over two millennia, one of the most common herbs prescribed in traditional Chinese medicine (TCM). ARS was developed as an antimalarial drug and currently belongs to the established standard treatments of malaria as a combination therapy worldwide. In addition to the antimalarial bioactivity of ARS, anticancer activities have been shown both in vitro and in vivo. Like other natural products, ARS acts in a multi-specific manner also against hematological malignancies. The chemical structure of ARS is a sesquiterpene lactone, which contains an endoperoxide bridge essential for activity. The main mechanism of action of ARS and its derivatives (artesunate, dihydroartemisinin, artemether) toward leukemia, multiple myeloma, and lymphoma cells comprises oxidative stress response, inhibition of proliferation, induction of various types of cell death as apoptosis, autophagy, ferroptosis, inhibition of angiogenesis, and signal transducers, as NF-κB, MYC, amongst others. Therefore, new pharmaceutically active compounds, dimers, trimers, and hybrid molecules, could enhance the existing therapeutic alternatives in combating hematologic malignancies. Owing to the high potency and good tolerance without side effects of ARS-type drugs, combination therapies with standard chemotherapies could be applied in the future after further clinical trials in hematological malignancies.
Collapse
Affiliation(s)
- R I Mancuso
- Hematology and Hemotherapy Center, University of Campinas, HEMOCENTRO UNICAMP, Campinas, São Paulo, Brazil
| | - M A Foglio
- Faculty of Pharmaceutical Science, University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | - S T Olalla Saad
- Hematology and Hemotherapy Center, University of Campinas, HEMOCENTRO UNICAMP, Campinas, São Paulo, Brazil.
| |
Collapse
|
12
|
Xu C, Zhang H, Mu L, Yang X. Artemisinins as Anticancer Drugs: Novel Therapeutic Approaches, Molecular Mechanisms, and Clinical Trials. Front Pharmacol 2020; 11:529881. [PMID: 33117153 PMCID: PMC7573816 DOI: 10.3389/fphar.2020.529881] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 09/16/2020] [Indexed: 12/26/2022] Open
Abstract
Artemisinin and its derivatives have shown broad-spectrum antitumor activities in vitro and in vivo. Furthermore, outcomes from a limited number of clinical trials provide encouraging evidence for their excellent antitumor activities. However, some problems such as poor solubility, toxicity and controversial mechanisms of action hamper their use as effective antitumor agents in the clinic. In order to accelerate the use of ARTs in the clinic, researchers have recently developed novel therapeutic approaches including developing novel derivatives, manufacturing novel nano-formulations, and combining ARTs with other drugs for cancer therapy. The related mechanisms of action were explored. This review describes ARTs used to induce non-apoptotic cell death containing oncosis, autophagy, and ferroptosis. Moreover, it highlights the ARTs-caused effects on cancer metabolism, immunosuppression and cancer stem cells and discusses clinical trials of ARTs used to treat cancer. The review provides additional insight into the molecular mechanism of action of ARTs and their considerable clinical potential.
Collapse
Affiliation(s)
- Cangcang Xu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, China
| | - Huihui Zhang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, China
| | - Lingli Mu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, China
| | - Xiaoping Yang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, China
| |
Collapse
|
13
|
Alven S, Aderibigbe BA. Nanoparticles Formulations of Artemisinin and Derivatives as Potential Therapeutics for the Treatment of Cancer, Leishmaniasis and Malaria. Pharmaceutics 2020; 12:E748. [PMID: 32784933 PMCID: PMC7466127 DOI: 10.3390/pharmaceutics12080748] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/23/2020] [Accepted: 04/28/2020] [Indexed: 12/15/2022] Open
Abstract
Cancer, malaria, and leishmaniasis remain the deadly diseases around the world although several strategies of treatment have been developed. However, most of the drugs used to treat the aforementioned diseases suffer from several pharmacological limitations such as poor pharmacokinetics, toxicity, drug resistance, poor bioavailability and water solubility. Artemisinin and its derivatives are antimalarial drugs. However, they also exhibit anticancer and antileishmanial activity. They have been evaluated as potential anticancer and antileishmanial drugs but their use is also limited by their poor water solubility and poor bioavailability. To overcome the aforementioned limitations associated with artemisinin and its derivatives used for the treatment of these diseases, they have been incorporated into nanoparticles. Several researchers incorporated this class of drugs into nanoparticles resulting in enhanced therapeutic outcomes. Their potential efficacy for the treatment of parasitic infections such as malaria and leishmaniasis and chronic diseases such as cancer has been reported. This review article will be focused on the nanoparticles formulations of artemisinin and derivatives for the treatment of cancer, malaria, and leishmaniasis and the biological outcomes (in vitro and in vivo).
Collapse
|
14
|
Tsamesidis I, Pério P, Pantaleo A, Reybier K. Oxidation of Erythrocytes Enhance the Production of Reactive Species in the Presence of Artemisinins. Int J Mol Sci 2020; 21:ijms21134799. [PMID: 32646002 PMCID: PMC7369783 DOI: 10.3390/ijms21134799] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/26/2020] [Accepted: 06/28/2020] [Indexed: 12/16/2022] Open
Abstract
In red blood cells, hemoglobin iron represents the most plausible candidate to catalyze artemisinin activation but the limited reactivity of iron bound to hemoglobin does not play in favor for its direct involvement. Denatured hemoglobin appears a more likely candidate for artemisinin redox activation because it is expected to contain reactive iron and it has been described to release free heme and/or iron in erythrocyte. The aim of our study is to investigate, using three different methods: fluorescence, electron paramagnetic resonance and liquid chromatography coupled to mass spectrometry, how increasing the level of accessible iron into the red blood cells can enhance the reactive oxygen species (ROS) production derived from artemisinin. The over-increase of iron was achieved using phenylhydrazine, a strong oxidant that causes oxidative stress within erythrocytes, resulting in oxidation of oxyhemoglobin and leading to the formation of methemoglobin, which is subsequently converted into irreversible hemichromes (iron (III) compounds). Our findings confirmed, using the iron III chelator, desferrioxamine, the indirect participation of iron (III) compounds in the activation process of artemisinins. Furthermore, in strong reducing conditions, the activation of artemisinin and the consequent production of ROS was enhanced. In conclusion, we demonstrate, through the measurement of intra-erythrocytic superoxide and hydrogen peroxide production using various methods, that artemisinin activation can be drastically enhanced by pre-oxidation of erythrocytes.
Collapse
Affiliation(s)
- Ioannis Tsamesidis
- Pharma-Dev UMR 152, Université de Toulouse, IRD, UPS, 31000 Toulouse, France; (P.P.); (K.R.)
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy;
- Correspondence:
| | - Pierre Pério
- Pharma-Dev UMR 152, Université de Toulouse, IRD, UPS, 31000 Toulouse, France; (P.P.); (K.R.)
| | - Antonella Pantaleo
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy;
| | - Karine Reybier
- Pharma-Dev UMR 152, Université de Toulouse, IRD, UPS, 31000 Toulouse, France; (P.P.); (K.R.)
| |
Collapse
|
15
|
Karol SE, Pui CH. Personalized therapy in pediatric high-risk B-cell acute lymphoblastic leukemia. Ther Adv Hematol 2020; 11:2040620720927575. [PMID: 32537116 PMCID: PMC7268159 DOI: 10.1177/2040620720927575] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022] Open
Abstract
Although cure rates for pediatric acute lymphoblastic leukemia (ALL) have now risen to more than 90%, subsets of patients with high-risk features continue to experience high rates of treatment failure and relapse. Recent work in minimal residual disease stratification and leukemia genomics have increased the ability to identify and classify these high-risk patients. In this review, we discuss this work to identify and classify patients with high-risk ALL. Novel therapeutics, which may have the potential to improve outcomes for these patients, are also discussed.
Collapse
Affiliation(s)
- Seth E Karol
- Department of Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl., Mail Stop 260, Memphis, TN 38105, USA
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
16
|
Abstract
Artemisinin (ART) and its derivatives are one of the most important classes of antimalarial agents, originally derived from a Chinese medicinal plant called Artemisia annua L. Beyond their outstanding antimalarial and antischistosomal activities, ART and its derivatives also possess both in-vitro and in-vivo activities against various types of cancer. Their anticancer effects range from initiation of apoptotic cell death to inhibition of cancer proliferation, metastasis and angiogenesis, and even modulation of the cell signal transduction pathway. This review provides a comprehensive update on ART and its derivatives, their mechanisms of action, and their synergistic effects with other chemicals in targeting leukemia cells. Combined with limited evidence of drug resistance and low toxicity profile, we conclude that ART and its derivatives, including dimers, trimers, and hybrids, might be a potential therapeutic alternative to current chemotherapies in combating leukemia, although more studies are necessary before they can be applied clinically.
Collapse
|
17
|
Li Y, Shan NN, Sui XH. Research Progress on Artemisinin and Its Derivatives against Hematological Malignancies. Chin J Integr Med 2020; 26:947-955. [PMID: 32048169 DOI: 10.1007/s11655-019-3207-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2019] [Indexed: 12/19/2022]
Abstract
Although current therapeutic methods against hematological malignancies are effective in the early stage, they usually lose their effectiveness because of the development of drug resistances. Seeking new drugs with significant therapeutic effects is one of the current research hotspots. Artemisinin, an extract from the plant Artemisia annua Linne, and its derivatives have excellent antimalarial effects in clinical applications as well as excellent safety. Recent studies have documented that artemisinin and its derivatives (ARTs) also have significant effects against multiple types of tumours, including hematological malignancies. This review focuses on the latest research achievements of ARTs in the treatment of hematological malignancies as well as its mechanisms and future applications. The mechanisms of ARTs against different types of hematological malignancies mainly include cell cycle arrest, induction autophagy and apoptosis, inhibition of angiogenesis, production of reactive oxygen species, and induction of differentiation. Additionally, the review also summarizes the anticancer effects of ARTs in many drug-resistant hematological malignancies and its synergistic effects with other drugs.
Collapse
Affiliation(s)
- Ying Li
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China.
| | - Ning-Ning Shan
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China
| | - Xiao-Hui Sui
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China
| |
Collapse
|
18
|
Song X, Liu Z, Yu Z. EGFR Promotes the Development of Triple Negative Breast Cancer Through JAK/STAT3 Signaling. Cancer Manag Res 2020; 12:703-717. [PMID: 32099467 PMCID: PMC6996555 DOI: 10.2147/cmar.s225376] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/07/2019] [Indexed: 12/28/2022] Open
Abstract
Purpose To investigate the role of EGFR and STAT3 in breast cancer development and progression. Methods Through bioinformatics analysis differently expressed genes (DEGs) including EGFR and STAT3 were identified in breast cancer tissue. QRT-PCR and Western blot analysis were used to investigate EGFR and STAT3 levels in breast cancer tissues and cells. The influence of EGFR and STAT3 on the breast cancer cell proliferation (CCK-8 assay, clone formation assays), migration (wound healing assays) and invasion (transwell assays) were investigated. The influence of EGFR on breast cancer in vivo was examined by Nude mouse transplantation tumor experiments and immunohistochemistry (IHC) staining. The effects of EGFR on breast cancer signaling were assessed via Western blot. Results Both EGFR and p-STAT3 were up-regulated in breast cancer tissues and cell lines. EGFR expression was positively associated with p-STAT3. Moreover, EGFR and p-STAT3 activity enhanced the proliferation and invasion of tumor cells. Breast cancer cell growth was dramatically inhibited by EGFR silencing in vivo. Conclusion EGFR promotes breast cancer progression via STAT3 phosphorylation and JAK/STAT3 signaling.
Collapse
Affiliation(s)
- Xiang Song
- Department of Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong, People's Republic of China
| | - Zhaoyun Liu
- Department of Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong, People's Republic of China.,School of Medicine, Shandong University, Jinan, People's Republic of China
| | - Zhiyong Yu
- Department of Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong, People's Republic of China
| |
Collapse
|
19
|
Yang L, Qiu Q, Tang M, Wang F, Yi Y, Yi D, Yang Z, Zhu Z, Zheng S, Yang J, Pei H, Zheng L, Chen Y, Gou L, Luo L, Deng X, Ye H, Hu Y, Niu T, Chen L. Purinostat Mesylate Is a Uniquely Potent and Selective Inhibitor of HDACs for the Treatment of BCR-ABL-Induced B-Cell Acute Lymphoblastic Leukemia. Clin Cancer Res 2019; 25:7527-7539. [PMID: 31439580 DOI: 10.1158/1078-0432.ccr-19-0516] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 06/30/2019] [Accepted: 08/13/2019] [Indexed: 02/05/2023]
Abstract
PURPOSE This study was to perform preclinical evaluation of a novel class I and IIb HDAC-selective inhibitor, purinostat mesylate, for the treatment of Ph+ B-cell acute lymphoblastic leukemia (B-ALL). EXPERIMENTAL DESIGN Biochemical assays were used to test enzymatic activity inhibition of purinostat mesylate. Ph+ leukemic cell lines and patient cells were used to evaluate purinostat mesylate activity in vitro. BL-2 secondary transplantation Ph+ B-ALL mouse model was used to validate its efficacy, mechanism, and pharmacokinetics properties in vivo. BCR-ABL(T315I)-induced primary B-ALL mouse model and PDX mouse model derived from relapsed Ph+ B-ALL patient post TKI treatment were used to determine the antitumor effect of purinostat mesylate for refractory or relapsed Ph+ B-ALL. Long-term toxicity and hERG blockade assays were used to safety evaluation of purinostat mesylate. RESULTS Purinostat mesylate, a class I and IIb HDAC highly selective inhibitor, exhibited robust antitumor activity in hematologic cancers. Purinostat mesylate at low nanomolar concentration induced apoptosis, and downregulated BCR-ABL and c-MYC expression in Ph+ leukemia cell lines and primary Ph+ B-ALL cells from relapsed patients. Purinostat mesylate efficiently attenuated Ph+ B-ALL progression and significantly prolonged the survival both in BL-2 secondary transplantation model with clinical patient symptoms of Ph+ B-ALL, BCR-ABL(T315I)-induced primary B-ALL mouse model, and PDX model derived from patients with relapsed Ph+ B-ALL post TKI treatment. In addition, purinostat mesylate possesses favorable pharmacokinetics and low toxicity properties. CONCLUSIONS Purinostat mesylate provides a new therapeutic strategy for patients with Ph+ B-ALL, including those who relapse after TKI treatment.
Collapse
Affiliation(s)
- Linyu Yang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Qiang Qiu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Minghai Tang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Fang Wang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Yuyao Yi
- Department of Hematology and Research Laboratory of Hematology, West China Hospital of Sichuan University, Chengdu, China
| | - Dongni Yi
- Department of Hematology and Research Laboratory of Hematology, West China Hospital of Sichuan University, Chengdu, China
| | - Zhuang Yang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Zejiang Zhu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Shoujun Zheng
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Jianhong Yang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Heying Pei
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Li Zheng
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Yong Chen
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Liping Gou
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Liya Luo
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Xing Deng
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Haoyu Ye
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Yiguo Hu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China.
| | - Ting Niu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China. .,Department of Hematology and Research Laboratory of Hematology, West China Hospital of Sichuan University, Chengdu, China
| | - Lijuan Chen
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|
20
|
Liu X, Cao J, Huang G, Zhao Q, Shen J. Biological Activities of Artemisinin Derivatives Beyond Malaria. Curr Top Med Chem 2019; 19:205-222. [DOI: 10.2174/1568026619666190122144217] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 11/19/2018] [Accepted: 11/21/2018] [Indexed: 12/26/2022]
Abstract
Artemisinin is isolated from Artemisia annua L. with peroxide-containing sesquiterpene lactone structure. Because of its unique structural characteristics and promising anticancer, antivirus activities, it has recently received increasing attention. The aim of this review is to summarize recent discoveries of artemisinin's novel derivatives with new pharmaceutical effects beyond malaria with a focus on its antitumor and antivirus activity, as well as potential results of combination therapy with other clinical drugs.
Collapse
Affiliation(s)
- Xiaoyan Liu
- CAS Key Laboratory for Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jianguo Cao
- College of Life and Environmental Sciences, Shanghai Normal University, Shanghai, 201418, China
| | - Guozheng Huang
- College of Life and Environmental Sciences, Shanghai Normal University, Shanghai, 201418, China
| | - Qingjie Zhao
- CAS Key Laboratory for Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jingshan Shen
- CAS Key Laboratory for Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
21
|
Liu X, Zhang Y, Huang W, Luo J, Li Y, Tan W, Zhang A. Development of high potent and selective Bcl-2 inhibitors bearing the structural elements of natural product artemisinin. Eur J Med Chem 2018; 159:149-165. [PMID: 30278333 DOI: 10.1016/j.ejmech.2018.09.059] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/23/2018] [Accepted: 09/24/2018] [Indexed: 11/24/2022]
Abstract
By taking advantage of the apoptosis-inducing capacity of artemisinin derivatives, we developed several series of compounds by merging the basic structural elements of the natural product artemisinin into the P2 interaction pocket of the clinically prescribed Bcl-2 inhibitor venetoclax. Most of the new compounds displayed improved biochemical potency against Bcl-2 and high selectivity over Bcl-xL. Specifically, compounds 27c and 34c were found to be the most potent with IC50 values less than 2.0 nM. Unfortunately, these compounds only showed moderate antiproliferative effects against Bcl-2 dependent cells. Though further structural optimization is needed to improve the cellular absorptive permeability, the current approach represents an alternative strategy to develop novel Bcl-2 inhibitors with greater selectivity over Bcl-xL, which is related to the off-target adverse effects of venetoclax.
Collapse
Affiliation(s)
- Xiaohua Liu
- CAS Key Laboratory of Receptor Research, The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yu Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Wenjing Huang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Jia Luo
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yang Li
- CAS Key Laboratory of Receptor Research, The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, 201203, China
| | - Wenfu Tan
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China.
| | - Ao Zhang
- CAS Key Laboratory of Receptor Research, The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, 201203, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
22
|
Zhang Y, Xu G, Zhang S, Wang D, Saravana Prabha P, Zuo Z. Antitumor Research on Artemisinin and Its Bioactive Derivatives. NATURAL PRODUCTS AND BIOPROSPECTING 2018; 8:303-319. [PMID: 29633188 PMCID: PMC6102173 DOI: 10.1007/s13659-018-0162-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/27/2018] [Indexed: 05/02/2023]
Abstract
Cancer is the leading cause of human death which seriously threatens human life. The antimalarial drug artemisinin and its derivatives have been discovered with considerable anticancer properties. Simultaneously, a variety of target-selective artemisinin-related compounds with high efficiency have been discovered. Many researches indicated that artemisinin-related compounds have cytotoxic effects against a variety of cancer cells through pleiotropic effects, including inhibiting the proliferation of tumor cells, promoting apoptosis, inducing cell cycle arrest, disrupting cancer invasion and metastasis, preventing angiogenesis, mediating the tumor-related signaling pathways, and regulating tumor microenvironment. More importantly, artemisinins demonstrated minor side effects to normal cells and manifested the ability to overcome multidrug-resistance which is widely observed in cancer patients. Therefore, we concentrated on the new advances and development of artemisinin and its derivatives as potential antitumor agents in recent 5 years. It is our hope that this review could be helpful for further exploration of novel artemisinin-related antitumor agents.
Collapse
Affiliation(s)
- Yunqin Zhang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Guowei Xu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Shuqun Zhang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Dong Wang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - P Saravana Prabha
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Zhili Zuo
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China.
- Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming, 650201, Yunnan, China.
| |
Collapse
|
23
|
Dihydroartemisinin suppresses STAT3 signaling and Mcl-1 and Survivin expression to potentiate ABT-263-induced apoptosis in Non-small Cell Lung Cancer cells harboring EGFR or RAS mutation. Biochem Pharmacol 2018; 150:72-85. [PMID: 29360439 DOI: 10.1016/j.bcp.2018.01.031] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 01/17/2018] [Indexed: 12/22/2022]
Abstract
Non-small cell lung cancer (NSCLC) is the most common malignancy worldwide. A significant fraction of NSCLC carries activating mutations in epidermal growth factor receptor (EGFR) or RAS oncogene. Dihydroartemisinin (DHA) is a semisynthetic derivative of the herbal antimalarial drug artemisinin that has been recently reported to exhibit anti-cancer activity. To develop new therapeutic strategies for NSCLC, we investigated the interactions between DHA and ABT-263 in NSCLC cells harboring EGFR or RAS mutation. Our data indicated that DHA synergized with ABT-263 to trigger Bax-dependent apoptosis in NSCLC cells in culture. DHA treatment antagonized ABT-263-induced Mcl-1 upregulation and sensitized NSCLC cells to ABT-263-triggered apoptosis. Additionally, DHA treatment caused downregulation of Survivin and upregulation of Bim, which also contribute to cotreatment-induced cytotoxicity. Moreover, DHA effectively suppressed STAT3 phosphorylation, and STAT3 inactivation resulted in the downregulation of Mcl-1 and Survivin, functioning to enhance ABT-263-induced cytotoxicity. Finally, cotreatment of DHA and ABT-263 significantly inhibited xenograft growth in nude mice. Together, DHA effectively inhibits STAT3 activity and modulates expression of Mcl-1, Survivin and Bim, thereby synergizing with ABT-263 to trigger apoptosis in NSCLC cells harboring EGFR or RAS mutation. Our data provide a novel therapeutic strategy for EGFR or RAS mutant NSCLC treatment.
Collapse
|