1
|
Kenmogne VL, Takundwa MM, Nweke EE, Monchusi B, Dube P, Maher H, Du Toit J, Philip-Cherian V, Fru PN, Thimiri Govinda Raj DB. The first-in-Africa ex vivo drug sensitivity testing platform identifies novel drug combinations for South African leukaemia patient cohort. Sci Rep 2025; 15:9160. [PMID: 40097584 PMCID: PMC11914478 DOI: 10.1038/s41598-025-93634-w] [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: 06/16/2024] [Accepted: 03/06/2025] [Indexed: 03/19/2025] Open
Abstract
In South Africa, leukemia remains a major health concern, posing significant challenges in treatment due to its varied subtypes. There is an unmet need for a testing pipeline that can identify drug effects on patient samples in an ex-vivo setting. Using the pilot study with South African patient samples, this paper reports the development of a drug-sensitivity testing pipeline for studying the drug effects in leukemia patient-derived cells. Forty-one (41) patients with Acute myeloid leukemia (AML) (n = 7), Chronic myelogenous leukemia (CML) (n = 30), and Chronic lymphocytic leukemia (CLL) (n = 4) were recruited for this study. Thirty (30) FDA-approved drugs were utilized for single drug sensitivity screening (DSS) on leukemia patient-derived cells with drug concentrations (1-1000 nM). The single DSS showed a distinct sensitivity pattern with different profiles among patients of the same subtype, confirming the need for precision therapy. This study observed irinotecan, used in solid tumour treatment, demonstrated efficacy in PBMCs in many patient samples compared to conventional leukemia drugs such as nilotinib. For drug combination studies, ten clinically relevant drugs were selected and tested based on the results of single drug sensitivity tests. This pilot study marks a crucial stride towards revolutionizing leukemia treatment in South Africa through an innovative ex vivo drug sensitivity testing platform. This pioneering initiative forms the basis for tailored and effective treatment options holding promise for more personalized treatment. Further exploration and validation of these findings could significantly contribute to cancer precision medicine efforts in South Africa.
Collapse
MESH Headings
- Humans
- South Africa
- Female
- Male
- Middle Aged
- Pilot Projects
- Adult
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Aged
- Drug Screening Assays, Antitumor/methods
- Leukemia, Myeloid, Acute/drug therapy
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Irinotecan/pharmacology
- Irinotecan/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Leukemia/drug therapy
- Leukemia/pathology
- Cohort Studies
- Pyrimidines
Collapse
Affiliation(s)
- V L Kenmogne
- Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future Production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - M M Takundwa
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future Production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - E E Nweke
- Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - B Monchusi
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future Production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - P Dube
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future Production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - H Maher
- Wits Donald Gordon Medical Centre, Johannesburg, South Africa
| | - J Du Toit
- Wits Donald Gordon Medical Centre, Johannesburg, South Africa
| | - V Philip-Cherian
- Department of Haematology, Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa
| | - P N Fru
- Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - D B Thimiri Govinda Raj
- Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa.
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future Production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa.
| |
Collapse
|
2
|
Fatima I, Ahmad R, Barman S, Gowrikumar S, Pravoverov K, Primeaux M, Fisher KW, Singh AB, Dhawan P. Albendazole inhibits colon cancer progression and therapy resistance by targeting ubiquitin ligase RNF20. Br J Cancer 2024; 130:1046-1058. [PMID: 38278978 PMCID: PMC10951408 DOI: 10.1038/s41416-023-02570-x] [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: 06/29/2023] [Revised: 12/08/2023] [Accepted: 12/21/2023] [Indexed: 01/28/2024] Open
Abstract
BACKGROUND The repurposing of FDA-approved drugs for anti-cancer therapies is appealing due to their established safety profiles and pharmacokinetic properties and can be quickly moved into clinical trials. Cancer progression and resistance to conventional chemotherapy remain the key hurdles in improving the clinical management of colon cancer patients and associated mortality. METHODS High-throughput screening (HTS) was performed using an annotated library of 1,600 FDA-approved drugs to identify drugs with strong anti-CRC properties. The candidate drug exhibiting most promising inhibitory effects in in-vitro studies was tested for its efficacy using in-vivo models of CRC progression and chemoresistance and patient derived organoids (PTDOs). RESULTS Albendazole, an anti-helminth drug, demonstrated the strongest inhibitory effects on the tumorigenic potentials of CRC cells, xenograft tumor growth and organoids from mice. Also, albendazole sensitized the chemoresistant CRC cells to 5-fluorouracil (5-FU) and oxaliplatin suggesting potential to treat chemoresistant CRC. Mechanistically, Albendazole treatment modulated the expression of RNF20, to promote apoptosis in CRC cells by delaying the G2/M phase and suppressing anti-apoptotic-Bcl2 family transcription. CONCLUSIONS Albendazole, an FDA approved drug, carries strong therapeutic potential to treat colon cancers which are aggressive and potentially resistant to conventional chemotherapeutic agents. Our findings also lay the groundwork for further clinical testing.
Collapse
Affiliation(s)
- Iram Fatima
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rizwan Ahmad
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Susmita Barman
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Saiprasad Gowrikumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kristina Pravoverov
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mark Primeaux
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kurt W Fisher
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Amar B Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
- Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
- VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA.
| |
Collapse
|
3
|
Pooler DB, Ness DB, Danilov AV, Labrie BM, Tosteson TD, Eastman A, Lewis LD, Lansigan F. A phase I trial of BNC105P and ibrutinib in patients with relapsed/refractory chronic lymphocytic leukemia. EJHAEM 2022; 3:1445-1448. [PMID: 36467840 PMCID: PMC9713021 DOI: 10.1002/jha2.543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 06/01/2023]
Affiliation(s)
- Darcy B. Pooler
- Sections of Clinical Pharmacology and Hematology OncologyDepartment of MedicineGeisel School of Medicine at Dartmouthand Dartmouth‐Hitchcock Medical CenterLebanonNew HampshireUSA
- The Dartmouth Cancer Center at Dartmouth‐Hitchcock Medical CenterLebanonNew HampshireUSA
| | - Dylan B. Ness
- Sections of Clinical Pharmacology and Hematology OncologyDepartment of MedicineGeisel School of Medicine at Dartmouthand Dartmouth‐Hitchcock Medical CenterLebanonNew HampshireUSA
- The Dartmouth Cancer Center at Dartmouth‐Hitchcock Medical CenterLebanonNew HampshireUSA
| | - Alexey V. Danilov
- Department of Hematology and Hematopoietic Cell TransplantationCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Bridget M. Labrie
- Sections of Clinical Pharmacology and Hematology OncologyDepartment of MedicineGeisel School of Medicine at Dartmouthand Dartmouth‐Hitchcock Medical CenterLebanonNew HampshireUSA
| | - Tor D. Tosteson
- The Dartmouth Cancer Center at Dartmouth‐Hitchcock Medical CenterLebanonNew HampshireUSA
- Department of Biomedical Data ScienceGeisel School of Medicine at DartmouthHanoverNew HampshireUSA
| | - Alan Eastman
- The Dartmouth Cancer Center at Dartmouth‐Hitchcock Medical CenterLebanonNew HampshireUSA
- Department of Molecular and Systems BiologyGeisel School of Medicine at DartmouthHanoverNew HampshireUSA
| | - Lionel D. Lewis
- Sections of Clinical Pharmacology and Hematology OncologyDepartment of MedicineGeisel School of Medicine at Dartmouthand Dartmouth‐Hitchcock Medical CenterLebanonNew HampshireUSA
- The Dartmouth Cancer Center at Dartmouth‐Hitchcock Medical CenterLebanonNew HampshireUSA
| | - Frederick Lansigan
- Sections of Clinical Pharmacology and Hematology OncologyDepartment of MedicineGeisel School of Medicine at Dartmouthand Dartmouth‐Hitchcock Medical CenterLebanonNew HampshireUSA
- The Dartmouth Cancer Center at Dartmouth‐Hitchcock Medical CenterLebanonNew HampshireUSA
| |
Collapse
|
4
|
Townsend PA, Kozhevnikova MV, Cexus ONF, Zamyatnin AA, Soond SM. BH3-mimetics: recent developments in cancer therapy. J Exp Clin Cancer Res 2021; 40:355. [PMID: 34753495 PMCID: PMC8576916 DOI: 10.1186/s13046-021-02157-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/26/2021] [Indexed: 01/11/2023] Open
Abstract
The hopeful outcomes from 30 years of research in BH3-mimetics have indeed served a number of solid paradigms for targeting intermediates from the apoptosis pathway in a variety of diseased states. Not only have such rational approaches in drug design yielded several key therapeutics, such outputs have also offered insights into the integrated mechanistic aspects of basic and clinical research at the genetics level for the future. In no other area of medical research have the effects of such work been felt, than in cancer research, through targeting the BAX-Bcl-2 protein-protein interactions. With these promising outputs in mind, several mimetics, and their potential therapeutic applications, have also been developed for several other pathological conditions, such as cardiovascular disease and tissue fibrosis, thus highlighting the universal importance of the intrinsic arm of the apoptosis pathway and its input to general tissue homeostasis. Considering such recent developments, and in a field that has generated so much scientific interest, we take stock of how the broadening area of BH3-mimetics has developed and diversified, with a focus on their uses in single and combined cancer treatment regimens and recently explored therapeutic delivery methods that may aid the development of future therapeutics of this nature.
Collapse
Affiliation(s)
- Paul A Townsend
- University of Surrey, Guildford, UK.
- Sechenov First Moscow State Medical University, Moscow, Russian Federation.
- University of Manchester, Manchester, UK.
| | - Maria V Kozhevnikova
- University of Surrey, Guildford, UK
- Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | | | - Andrey A Zamyatnin
- University of Surrey, Guildford, UK
- Sechenov First Moscow State Medical University, Moscow, Russian Federation
- Lomonosov Moscow State University, Moscow, Russian Federation
- Sirius University of Science and Technology, Sochi, Russian Federation
| | - Surinder M Soond
- University of Surrey, Guildford, UK.
- Sechenov First Moscow State Medical University, Moscow, Russian Federation.
| |
Collapse
|
5
|
Fermaintt CS, Takahashi-Ruiz L, Liang H, Mooberry SL, Risinger AL. Eribulin Activates the cGAS-STING Pathway via the Cytoplasmic Accumulation of Mitochondrial DNA. Mol Pharmacol 2021; 100:309-318. [PMID: 34312217 PMCID: PMC8626644 DOI: 10.1124/molpharm.121.000297] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/14/2021] [Indexed: 11/23/2022] Open
Abstract
Microtubule-targeting agents (MTAs), including both microtubule stabilizers and destabilizers are highly effective chemotherapeutic drugs used in the treatment of solid tumors and hematologic malignancies. In addition to the shared ability of all MTAs to block cell cycle progression, growing evidence shows that different agents of this class can also have mechanistically distinct effects on nonmitotic microtubule-dependent cellular processes, including cellular signaling and transport. Herein, we test the biologic hypothesis that MTAs used in the treatment of triple-negative breast cancer (TNBC) can differentially affect innate immune signaling pathways independent of their antimitotic effects. Our data demonstrate that the microtubule destabilizer eribulin, but not the microtubule stabilizer paclitaxel, induces cGAS-STING-dependent expression of interferon-β in both myeloid and TNBC cells. Activation of the cGAS-STING pathway by eribulin was further found to be mediated by the accumulation of cytoplasmic mitochondrial DNA. Together, these findings provide mechanistic insight into how eribulin can induce innate immune signaling independent of its antimitotic or cytotoxic effects. SIGNIFICANCE STATEMENT: Microtubule-targeting agents (MTAs) are often used in the treatment of breast cancer and have been used in combination with immune checkpoint inhibitors to improve efficacy. Although all clinically approved MTAs share an antimitotic mechanism of action, their distinct effects on interphase microtubules can promote differential downstream signaling consequences. This work shows that the microtubule destabilizer eribulin, but not the microtubule stabilizer paclitaxel, activates the cGAS-STING innate immune signaling pathway through the accumulation of mitochondrial DNA in the cytoplasm.
Collapse
Affiliation(s)
- Charles S Fermaintt
- Department of Pharmacology (C.S.F., L.T.-R., H.L., S.L.M., A.L.R.), and Mays Cancer Center (C.S.F., S.L.M., A.L.R.), University of Texas Health Science Center San Antonio, San Antonio, Texas
| | - Leila Takahashi-Ruiz
- Department of Pharmacology (C.S.F., L.T.-R., H.L., S.L.M., A.L.R.), and Mays Cancer Center (C.S.F., S.L.M., A.L.R.), University of Texas Health Science Center San Antonio, San Antonio, Texas
| | - Huiyun Liang
- Department of Pharmacology (C.S.F., L.T.-R., H.L., S.L.M., A.L.R.), and Mays Cancer Center (C.S.F., S.L.M., A.L.R.), University of Texas Health Science Center San Antonio, San Antonio, Texas
| | - Susan L Mooberry
- Department of Pharmacology (C.S.F., L.T.-R., H.L., S.L.M., A.L.R.), and Mays Cancer Center (C.S.F., S.L.M., A.L.R.), University of Texas Health Science Center San Antonio, San Antonio, Texas
| | - April L Risinger
- Department of Pharmacology (C.S.F., L.T.-R., H.L., S.L.M., A.L.R.), and Mays Cancer Center (C.S.F., S.L.M., A.L.R.), University of Texas Health Science Center San Antonio, San Antonio, Texas
| |
Collapse
|
6
|
Mallick DJ, Eastman A. AT101 [(-)-Gossypol] Selectively Inhibits MCL1 and Sensitizes Carcinoma to BH3 Mimetics by Inducing and Stabilizing NOXA. Cancers (Basel) 2020; 12:E2298. [PMID: 32824203 PMCID: PMC7464284 DOI: 10.3390/cancers12082298] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/04/2020] [Accepted: 08/12/2020] [Indexed: 12/17/2022] Open
Abstract
Anti-apoptotic BCL2 proteins are important targets for cancer therapy as cancers depend on their activity for survival. Direct inhibitors of MCL1 have entered clinical trials, although their efficacy may be limited by toxicity. An alternative approach may be to induce the pro-apoptotic protein NOXA which selectively inhibits MCL1 in cells. Many compounds originally proposed as inhibitors of the BCL2 family were subsequently found to induce the pro-apoptotic protein NOXA through the unfolded protein response. In the present study, we compared various putative BH3 mimetics across a panel of carcinoma cell lines and measured expression of NOXA protein and mRNA, as well as the kinetics of NOXA induction. We found that AT101 [(-)-gossypol] induces high levels of NOXA in carcinoma cell lines yet cells survive. When combined with an appropriate BCL2 or BCL-XL inhibitor, NOXA-dependent sensitization occurs. NOXA protein continues to accumulate for many hours after AT101 is removed, providing a window for administering these combinations. As MCL1 promotes drug resistance and overall survival, we propose that NOXA induction is an alternative therapeutic strategy to target MCL1 and either kill cancer cells that are dependent on MCL1 or sensitize cancer cells to other BCL2 inhibitors.
Collapse
Affiliation(s)
- David J. Mallick
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA;
| | - Alan Eastman
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA;
- Department of Molecular and Systems Biology, and Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| |
Collapse
|
7
|
Masilamani AP, Dettmer-Monaco V, Monaco G, Cathomen T, Kuckuck I, Schultze-Seemann S, Huber N, Wolf P. An Anti-PSMA Immunotoxin Reduces Mcl-1 and Bcl2A1 and Specifically Induces in Combination with the BAD-Like BH3 Mimetic ABT-737 Apoptosis in Prostate Cancer Cells. Cancers (Basel) 2020; 12:cancers12061648. [PMID: 32580291 PMCID: PMC7352695 DOI: 10.3390/cancers12061648] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 12/25/2022] Open
Abstract
Background: Upregulation of anti-apoptotic Bcl-2 proteins in advanced prostate cancer leads to therapeutic resistance by prevention of cell death. New therapeutic approaches aim to target the Bcl-2 proteins for the restoration of apoptosis. Methods: The immunotoxin hD7-1(VL-VH)-PE40 specifically binds to the prostate specific membrane antigen (PSMA) on prostate cancer cells and inhibits protein biosynthesis. It was tested with respect to its effects on the expression of anti-apoptotic Bcl-2 proteins. Combination with the BAD-like mimetic ABT-737 was examined on prostate cancer cells and 3D spheroids and in view of tumor growth and survival in the prostate cancer SCID mouse xenograft model. Results: The immunotoxin led to a specific inhibition of Mcl-1 and Bcl2A1 expression in PSMA expressing target cells. Its combination with ABT-737, which inhibits Bcl-2, Bcl-xl, and Bcl-w, led to an induction of the intrinsic apoptotic pathway and to a synergistic cytotoxicity in prostate cancer cells and 3D spheroids. Furthermore, combination therapy led to a significantly prolonged survival of mice bearing prostate cancer xenografts based on an inhibition of tumor growth. Conclusion: The combination therapy of anti-PSMA immunotoxin plus ABT-737 represents the first tumor-specific therapeutic approach on the level of Bcl-2 proteins for the induction of apoptosis in prostate cancer.
Collapse
Affiliation(s)
- Anie P. Masilamani
- Department of Urology, Medical Center—University of Freiburg, 79106 Freiburg, Germany; (A.P.M.); (I.K.); (S.S.-S.); (N.H.)
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (V.D.-M.); (G.M.); (T.C.)
| | - Viviane Dettmer-Monaco
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (V.D.-M.); (G.M.); (T.C.)
- Institute for Transfusion Medicine and Gene Therapy, Medical Center—University of Freiburg, 79106 Freiburg, Germany
- Center for Chronic Immunodeficiency, University of Freiburg, 79106 Freiburg, Germany
| | - Gianni Monaco
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (V.D.-M.); (G.M.); (T.C.)
- Institute for Transfusion Medicine and Gene Therapy, Medical Center—University of Freiburg, 79106 Freiburg, Germany
- Center for Chronic Immunodeficiency, University of Freiburg, 79106 Freiburg, Germany
| | - Toni Cathomen
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (V.D.-M.); (G.M.); (T.C.)
- Institute for Transfusion Medicine and Gene Therapy, Medical Center—University of Freiburg, 79106 Freiburg, Germany
- Center for Chronic Immunodeficiency, University of Freiburg, 79106 Freiburg, Germany
| | - Irina Kuckuck
- Department of Urology, Medical Center—University of Freiburg, 79106 Freiburg, Germany; (A.P.M.); (I.K.); (S.S.-S.); (N.H.)
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (V.D.-M.); (G.M.); (T.C.)
| | - Susanne Schultze-Seemann
- Department of Urology, Medical Center—University of Freiburg, 79106 Freiburg, Germany; (A.P.M.); (I.K.); (S.S.-S.); (N.H.)
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (V.D.-M.); (G.M.); (T.C.)
| | - Nathalie Huber
- Department of Urology, Medical Center—University of Freiburg, 79106 Freiburg, Germany; (A.P.M.); (I.K.); (S.S.-S.); (N.H.)
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (V.D.-M.); (G.M.); (T.C.)
| | - Philipp Wolf
- Department of Urology, Medical Center—University of Freiburg, 79106 Freiburg, Germany; (A.P.M.); (I.K.); (S.S.-S.); (N.H.)
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (V.D.-M.); (G.M.); (T.C.)
- Correspondence: ; Tel.: +49-761-270-28921
| |
Collapse
|
8
|
Barreca M, Stathis A, Barraja P, Bertoni F. An overview on anti-tubulin agents for the treatment of lymphoma patients. Pharmacol Ther 2020; 211:107552. [PMID: 32305312 DOI: 10.1016/j.pharmthera.2020.107552] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/14/2020] [Indexed: 01/19/2023]
Abstract
Anti-tubulin agents constitute a large class of compounds with broad activity both in solid tumors and hematologic malignancies, due to the interference with microtubule dynamics. Since microtubules play crucial roles in the regulation of the mitotic spindles, the interference with their function usually leads to a block in cell division with arrest at the metaphase/anaphase junction of mitosis, followed to apoptosis. This explains the reason why tubulin-binding agents (TBAs) proved to be extremely active in patients with cancer. Several anti-tubulin agents are indicated in the treatment of patients with lymphomas both alone and in combination chemotherapy regimens. The article reviews the literature on classic and more recent anti-tubulin agents, providing an insight into their mechanisms of action and their use in the treatment of lymphoma.
Collapse
Affiliation(s)
- Marilia Barreca
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy; Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
| | - Anastasios Stathis
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland,; Faculty of Biomedical Sciences, USI, Lugano, Switzerland
| | - Paola Barraja
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland,; Oncology Institute of Southern Switzerland, Bellinzona, Switzerland,.
| |
Collapse
|
9
|
BH3 mimetic ABT-263 enhances the anticancer effects of apigenin in tumor cells with activating EGFR mutation. Cell Biosci 2019; 9:60. [PMID: 31367332 PMCID: PMC6651933 DOI: 10.1186/s13578-019-0322-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 07/05/2019] [Indexed: 01/11/2023] Open
Abstract
Background Mutated epidermal growth factor receptor (EGFR) is one of the most successful targets in cancer targeted therapy. While this treatment has benefited many patients with an activating EGFR mutation (EGFRm), almost all those who initially benefited will eventually develop acquired drug resistance (ADR) after a certain period of time. New therapeutic strategies need to be explored to treat EGFRm tumors and overcome or minimize this recurring ADR. Results Our data showed that apigenin alone has only mild inhibitory effects on EGFRm tumor cells. By drug screening, we found that ABT-263 can significantly enhance the antitumor activities of apigenin in tumor cells harbouring an activating EGFR mutation and AZD9291-resistant H1975 cells. Mechanistically, apigenin upregulated the expression of Noxa in EGFRm tumor cells by targeting the AKT-FoxO3a pathway, thereby synergizing with ABT-263 to suppress tumor cell growth and proliferation in vitro and in vivo. Conclusions Our study provides a rationale for the clinical application of the combination treatment of apigenin and BH3 mimetics in the treatment of EGFRm tumors. Electronic supplementary material The online version of this article (10.1186/s13578-019-0322-y) contains supplementary material, which is available to authorized users.
Collapse
|
10
|
Non-mitotic effect of albendazole triggers apoptosis of human leukemia cells via SIRT3/ROS/p38 MAPK/TTP axis-mediated TNF-α upregulation. Biochem Pharmacol 2019; 162:154-168. [DOI: 10.1016/j.bcp.2018.11.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 11/05/2018] [Indexed: 12/27/2022]
|
11
|
Eastman A. Improving anticancer drug development begins with cell culture: misinformation perpetrated by the misuse of cytotoxicity assays. Oncotarget 2018; 8:8854-8866. [PMID: 27750219 PMCID: PMC5352448 DOI: 10.18632/oncotarget.12673] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 10/12/2016] [Indexed: 11/25/2022] Open
Abstract
The high failure rate of anticancer drug discovery and development has consumed billions of dollars annually. While many explanations have been provided, I believe that misinformation arising from inappropriate cell-based screens has been completely over-looked. Most cell culture experiments are irrelevant to how drugs are subsequently administered to patients. Usually, drug development focuses on growth inhibition rather than cell killing. Drugs are selected based on continuous incubation of cells, then frequently administered to the patient as a bolus. Target identification and validation is often performed by gene suppression that inevitably mimics continuous target inhibition. Drug concentrations in vitro frequently far exceed in vivo concentrations. Studies of drug synergy are performed at sub-optimal concentrations. And the focus on a limited number of cell lines can misrepresent the potential efficacy in a patient population. The intent of this review is to encourage more appropriate experimental design and data interpretation, and to improve drug development in the area of cell-based assays. Application of these principles should greatly enhance the successful translation of novel drugs to the patient.
Collapse
Affiliation(s)
- Alan Eastman
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| |
Collapse
|
12
|
Opydo-Chanek M, Gonzalo O, Marzo I. Multifaceted anticancer activity of BH3 mimetics: Current evidence and future prospects. Biochem Pharmacol 2017; 136:12-23. [PMID: 28288819 DOI: 10.1016/j.bcp.2017.03.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 03/06/2017] [Indexed: 12/19/2022]
Abstract
BH3 mimetics are a novel class of anticancer agents designed to specifically target pro-survival proteins of the Bcl-2 family. Like endogenous BH3-only proteins, BH3 mimetics competitively bind to surface hydrophobic grooves of pro-survival Bcl-2 family members, counteracting their protective effects and thus facilitating apoptosis in cancer cells. Among the small-molecule BH3 mimetics identified, ABT-737 and its analogs, obatoclax as well as gossypol derivatives are the best characterized. The anticancer potential of these compounds applied as a single agent or in combination with chemotherapeutic drugs is currently being evaluated in preclinical studies and in clinical trials. In spite of promising results, the actual mechanisms of their anticancer action remain to be identified. Findings from preclinical studies point to additional activities of BH3 mimetics in cancer cells that are not connected with apoptosis induction. These off-target effects involve induction of autophagy and necrotic cell death as well as modulation of the cell cycle and multiple cell signaling pathways. For the optimization and clinical implementation of BH3 mimetics, a detailed understanding of their role as inhibitors of the pro-survival Bcl-2 proteins, but also of their possible additional effects is required. This review summarizes the most representative BH3 mimetic compounds with emphasis on their off-target effects. Based on the present knowledge on the multifaceted effects of BH3 mimetics on cancer cells, the commentary outlines the potential pitfalls and highlights the considerable promise for cancer treatment with BH3 mimetics.
Collapse
Affiliation(s)
- Małgorzata Opydo-Chanek
- Department of Experimental Hematology, Institute of Zoology, Jagiellonian University in Kraków, Poland.
| | - Oscar Gonzalo
- Department of Biochemistry, Molecular and Cell Biology, IIS, University of Zaragoza, Spain
| | - Isabel Marzo
- Department of Biochemistry, Molecular and Cell Biology, IIS, University of Zaragoza, Spain
| |
Collapse
|
13
|
Petrov P, Syrjänen R, Uchida T, Vainio O. Leucocyte protein Trojan, a possible regulator of apoptosis. APMIS 2016; 125:106-113. [PMID: 28028869 DOI: 10.1111/apm.12641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 10/27/2016] [Indexed: 11/28/2022]
Abstract
Trojan is a leucocyte-specific protein, cloned from chicken embryonic thymocyte cDNA library. The molecule is a type I transmembrane protein with an extracellular CCP domain, followed by two FN3 domains. Its cytoplasmic tail is predicted to possess a MAPK docking and a PKA phosphorylation sites. Trojan has been proposed to have an anti-apoptotic role based on its differential expression on developing thymocyte subpopulations. Using a chicken cell line, our in vitro studies showed that upon apoptosis induction, Trojan expression rises dramatically on the surface of surviving cells and gradually decreases towards its normal levels as cells recover. When sorted based on their expression levels of Trojan, cells with high expression appeared less susceptible to apoptotic induction than those bearing no or low levels of Trojan on their surface. The mechanism by which the molecule exerts its function is yet to be discovered. We found that cells overexpressing Trojan from a cDNA plasmid show elevated steady-state levels of intracellular calcium, suggesting the molecule is able to transmit cytoplasmic signals. The mechanistic nature of Trojan-induced signalling is a target of future investigation. In this article, we conducted a series of experiments that suggest Trojan as an anti-apoptotic regulator.
Collapse
Affiliation(s)
- Petar Petrov
- Research Unit of Biomedicine, Department of Medical Microbiology and Immunology, University of Oulu, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.,Nordlab Oulu, Oulu, Finland
| | - Riikka Syrjänen
- Research Unit of Biomedicine, Department of Medical Microbiology and Immunology, University of Oulu, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.,Nordlab Oulu, Oulu, Finland
| | - Tatsuya Uchida
- Research Unit of Biomedicine, Department of Medical Microbiology and Immunology, University of Oulu, Oulu, Finland
| | - Olli Vainio
- Research Unit of Biomedicine, Department of Medical Microbiology and Immunology, University of Oulu, Oulu, Finland.,Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.,Nordlab Oulu, Oulu, Finland
| |
Collapse
|
14
|
Bates D, Eastman A. Microtubule destabilising agents: far more than just antimitotic anticancer drugs. Br J Clin Pharmacol 2016; 83:255-268. [PMID: 27620987 DOI: 10.1111/bcp.13126] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 08/11/2016] [Accepted: 09/07/2016] [Indexed: 02/06/2023] Open
Abstract
Vinca alkaloids have been approved as anticancer drugs for more than 50 years. They have been classified as cytotoxic chemotherapy drugs that act during cellular mitosis, enabling them to target fast growing cancer cells. With the evolution of cancer drug development there has been a shift towards new "targeted" therapies to avoid the side effects and general toxicities of "cytotoxic chemotherapies" such as the vinca alkaloids. Due to their original classification, many have overlooked the fact that vinca alkaloids, taxanes and related drugs do have a specific molecular target: tubulin. They continue to be some of the most effective anticancer drugs, perhaps because their actions upon the microtubule network extend far beyond the ability to halt cells in mitosis, and include the induction of apoptosis at all phases of the cell cycle. In this review, we highlight the numerous cellular consequences of disrupting microtubule dynamics, expanding the textbook knowledge of microtubule destabilising agents and providing novel opportunities for their use in cancer therapy.
Collapse
Affiliation(s)
- Darcy Bates
- Department of Medicine, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Alan Eastman
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| |
Collapse
|
15
|
Kim LH, Shin JA, Jang B, Yang IH, Won DH, Jeong JH, Chung TH, Cho NP, Cho SD. Sorafenib potentiates ABT-737-induced apoptosis in human oral cancer cells. Arch Oral Biol 2016; 73:1-6. [PMID: 27632413 DOI: 10.1016/j.archoralbio.2016.08.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 08/14/2016] [Accepted: 08/30/2016] [Indexed: 11/15/2022]
Abstract
OBJECTIVE The mimetic BH3 ABT-737, a potent inhibitor of anti-apoptotic Bcl-2 family proteins, has potential as anti-cancer drug in many cancers. Recently, patients treated with ABT-737 have developed drug tolerance during cancer therapy. Therefore, we examined whether ABT-737 is effective in killing MC-3 and HSC-3 human oral cancer cells either alone or in combination with the oncogenic kinase inhibitor, sorafenib. DESIGN The potentiating activities of sorafenib in ABT-737-induced apoptosis were determined using trypan blue exclusion assay, DAPI staining, cell viability assay and Western blot analysis. RESULTS Combined use of ABT-737 and sorafenib synergistically suppressed cell viability and induced apoptosis compared with either compound individually. The combination of ABT-737 and sorafenib altered only Bax and Bak proteins and their activations, resulting in mitochondrial translocation of Bax from the cytosol. Additionally, combination treatment-mediated apoptosis may be correlated with ERK and STAT3 pathways. CONCLUSIONS These results suggest that sorafenib may effectively overcome ABT-737 resistance to apoptotic cell death, which can be a new potential chemotherapeutic strategy against human oral cancer.
Collapse
Affiliation(s)
- Lee-Han Kim
- Department of Oral Pathology, School of Dentistry, Institute of Biodegradable material, Institute of Oral Bioscience, Chonbuk National University, Jeonju 54986, Republic of Korea
| | - Ji-Ae Shin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Alkek Building for Biomedical Research, Houston, TX, 77030, USA
| | - Boonsil Jang
- Department of Oral Pathology, School of Dentistry, Institute of Biodegradable material, Institute of Oral Bioscience, Chonbuk National University, Jeonju 54986, Republic of Korea
| | - In-Hyoung Yang
- Department of Oral Pathology, School of Dentistry, Institute of Biodegradable material, Institute of Oral Bioscience, Chonbuk National University, Jeonju 54986, Republic of Korea
| | - Dong-Hoon Won
- Department of Oral Pathology, School of Dentistry, Institute of Biodegradable material, Institute of Oral Bioscience, Chonbuk National University, Jeonju 54986, Republic of Korea
| | - Joseph H Jeong
- Department of Urology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Tae-Ho Chung
- Department of Animal Resources Science, Joongbu University, Chungnam, 32713, Republic of Korea
| | - Nam-Pyo Cho
- Department of Oral Pathology, School of Dentistry, Institute of Biodegradable material, Institute of Oral Bioscience, Chonbuk National University, Jeonju 54986, Republic of Korea
| | - Sung-Dae Cho
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea.
| |
Collapse
|
16
|
Kothari A, Hittelman WN, Chambers TC. Cell Cycle-Dependent Mechanisms Underlie Vincristine-Induced Death of Primary Acute Lymphoblastic Leukemia Cells. Cancer Res 2016; 76:3553-61. [PMID: 27197148 DOI: 10.1158/0008-5472.can-15-2104] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 04/03/2016] [Indexed: 01/08/2023]
Abstract
Microtubule-targeting agents (MTA), such as the taxanes and vinca alkaloids, are used to treat a variety of cancers due to their ability to perturb microtubule dynamics. In cell culture, MTAs exert their anticancer effects primarily by causing mitotic arrest and cell death. However, accumulating indirect evidence suggests that MTAs may exert their cytotoxicity in human tumors by interfering with interphase microtubules. In this study, we sought to develop and characterize an experimental system in which to test the hypothesis that MTAs induce cell death during interphase. Primary adult acute lymphoblastic leukemia (ALL) cells treated with vincristine only weakly exhibited colocalization between mitotic and apoptotic markers and major characteristics of mitotic death, such as an increase in cells with 4N DNA content before the appearance of cells with <2N DNA content, suggesting a mixed response. Therefore, we separated ALL cells into distinct phases of the cell cycle by centrifugal elutriation, labeled cells with 5-ethynyl-2'-deoxyuridine (EdU), and then treated each population with vincristine. Cells isolated during G1 underwent cell death without evidence of EdU uptake, indicating that the cytotoxic effects of vincristine took place during G1 Conversely, cells isolated during S or G2-M phases underwent death following mitotic arrest. Thus, vincristine induces distinct death programs in primary ALL cells depending on cell-cycle phase, and cells in G1 are particularly susceptible to perturbation of interphase microtubules. Primary ALL cells may therefore provide a powerful model system in which to study the multimodal mechanisms underlying MTA-induced cell death. Cancer Res; 76(12); 3553-61. ©2016 AACR.
Collapse
Affiliation(s)
- Anisha Kothari
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Walter N Hittelman
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Timothy C Chambers
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas.
| |
Collapse
|
17
|
Bates D, Feris EJ, Danilov AV, Eastman A. Rapid induction of apoptosis in chronic lymphocytic leukemia cells by the microtubule disrupting agent BNC105. Cancer Biol Ther 2016; 17:291-9. [PMID: 26891146 DOI: 10.1080/15384047.2016.1139245] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Microtubule targeting agents, such as vinblastine, are usually thought to arrest cells in mitosis and subsequently induce apoptosis. However, they can also cause rapid induction of apoptosis in a cell-cycle phase independent manner. BNC105 is a novel vascular and microtubule disrupting drug that also induces apoptosis rapidly but with markedly increased potency compared to vinca alkaloids and combretastatin A4. BNC105 binds to the colchicine-binding site on tubulin resulting in activation of c-Jun N-terminal kinase (JNK), phosphorylation of ATF2, and induction of ATF3 and Noxa leading to acute apoptosis in chronic lymphocytic leukemia (CLL) cells. Apoptosis induced by BNC105 is dependent upon both JNK activation and Noxa induction. Normal leukocytes and one CLL sample also exhibited JNK activation but not Noxa induction and were resistant to BNC105. This study emphasizes the importance of Noxa and JNK for induction of apoptosis in CLL cells by microtubule targeting drugs, and highlights the potential of BNC105 as a potent therapeutic to treat haematopoietic malignancies.
Collapse
Affiliation(s)
- Darcy Bates
- a Department of Medicine , Geisel School of Medicine at Dartmouth , Lebanon , NH , USA.,c Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth , Lebanon , NH , USA
| | - Edmond J Feris
- b Department of Pharmacology and Toxicology , Geisel School of Medicine at Dartmouth , Lebanon , NH , USA.,c Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth , Lebanon , NH , USA
| | - Alexey V Danilov
- a Department of Medicine , Geisel School of Medicine at Dartmouth , Lebanon , NH , USA.,c Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth , Lebanon , NH , USA
| | - Alan Eastman
- b Department of Pharmacology and Toxicology , Geisel School of Medicine at Dartmouth , Lebanon , NH , USA.,c Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth , Lebanon , NH , USA
| |
Collapse
|
18
|
Natural compounds for pediatric cancer treatment. Naunyn Schmiedebergs Arch Pharmacol 2015; 389:131-49. [DOI: 10.1007/s00210-015-1191-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 11/08/2015] [Indexed: 12/13/2022]
|
19
|
Paiva C, Godbersen JC, Soderquist RS, Rowland T, Kilmarx S, Spurgeon SE, Brown JR, Srinivasa SP, Danilov AV. Cyclin-Dependent Kinase Inhibitor P1446A Induces Apoptosis in a JNK/p38 MAPK-Dependent Manner in Chronic Lymphocytic Leukemia B-Cells. PLoS One 2015; 10:e0143685. [PMID: 26606677 PMCID: PMC4659573 DOI: 10.1371/journal.pone.0143685] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 11/09/2015] [Indexed: 01/17/2023] Open
Abstract
CDK (cyclin-dependent kinase) inhibitors have shown remarkable activity in CLL, where its efficacy has been linked to inhibition of the transcriptional CDKs (7 and 9) and deregulation of RNA polymerase and short-lived pro-survival proteins such as MCL1. Furthermore, ER (endoplasmic reticulum) stress has been implicated in CDK inhibition in CLL. Here we conducted a pre-clinical study of a novel orally active kinase inhibitor P1446A in CLL B-cells. P1446A inhibited CDKs at nanomolar concentrations and induced rapid apoptosis of CLL cells in vitro, irrespective of chromosomal abnormalities or IGHV mutational status. Apoptosis preceded inactivation of RNA polymerase, and was accompanied by phosphorylation of stress kinases JNK (c-Jun N-terminal kinase) and p38 MAPK (mitogen-activated protein kinase). Pharmacologic inhibitors of JNK/p38 MAPK conferred protection from P1446A-mediated apoptosis. Treatment with P1446A led to a dramatic induction of NOXA in a JNK-dependent manner, and sensitized CLL cells to ABT-737, a BH3-mimetic. We observed concurrent activation of apoptosis stress-inducing kinase 1 (ASK1) and its interaction with inositol-requiring enzyme 1 (IRE1) and tumor necrosis factor receptor-associated factor 2 (TRAF2) in CLL cells treated with P1446A, providing insights into upstream regulation of JNK in this setting. Consistent with previous reports on limited functionality of ER stress mechanism in CLL cells, treatment with P1446A failed to induce an extensive unfolded protein response. This study provides rationale for additional investigations of P1446A in CLL.
Collapse
Affiliation(s)
- Cody Paiva
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States of America
| | | | | | - Taylor Rowland
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States of America
| | - Sumner Kilmarx
- Dartmouth College, Hanover, NH, United States of America
| | - Stephen E. Spurgeon
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States of America
| | - Jennifer R. Brown
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States of America
| | | | - Alexey V. Danilov
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States of America
- * E-mail:
| |
Collapse
|
20
|
Greene LM, Meegan MJ, Zisterer DM. Combretastatins: more than just vascular targeting agents? J Pharmacol Exp Ther 2015; 355:212-27. [PMID: 26354991 DOI: 10.1124/jpet.115.226225] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 08/25/2015] [Indexed: 01/23/2023] Open
Abstract
Several prodrugs of the naturally occurring combretastatins have undergone extensive clinical evaluation as vascular targeting agents (VTAs). Their increased selectivity toward endothelial cells together with their innate ability to rapidly induce vascular shutdown and inhibit tumor growth at doses up to 10-fold less than the maximum tolerated dose led to the clinical evaluation of combretastatins as VTAs. Tubulin is well established as the molecular target of the combretastatins and the vast majority of its synthetic derivatives. Furthermore, tubulin is a highly validated molecular target of many direct anticancer agents routinely used as front-line chemotherapeutics. The unique vascular targeting properties of the combretastatins have somewhat overshadowed their development as direct anticancer agents and the delineation of the various cell death pathways and anticancer properties associated with such chemotherapeutics. Moreover, the ongoing clinical trial of OXi4503 (combretastatin-A1 diphosphate) together with preliminary preclinical evaluation for the treatment of refractory acute myelogenous leukemia has successfully highlighted both the indirect and direct anticancer properties of combretastatins. In this review, we discuss the development of the combretastatins from nature to the clinic. The various mechanisms underlying combretastatin-induced cell cycle arrest, mitotic catastrophe, cell death, and survival are also reviewed in an attempt to further enhance the clinical prospects of this unique class of VTAs.
Collapse
Affiliation(s)
- Lisa M Greene
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (L.M.G., D.M.Z.), and School of Pharmacy and Pharmaceutical Sciences, Centre for Synthesis and Chemical Biology (M.J.M.), Trinity College Dublin, Dublin, Ireland
| | - Mary J Meegan
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (L.M.G., D.M.Z.), and School of Pharmacy and Pharmaceutical Sciences, Centre for Synthesis and Chemical Biology (M.J.M.), Trinity College Dublin, Dublin, Ireland
| | - Daniela M Zisterer
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (L.M.G., D.M.Z.), and School of Pharmacy and Pharmaceutical Sciences, Centre for Synthesis and Chemical Biology (M.J.M.), Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
21
|
Bates DJP, Lewis LD, Eastman A, Danilov AV. Vincristine activates c-Jun N-terminal kinase in chronic lymphocytic leukaemia in vivo. Br J Clin Pharmacol 2015; 80:493-501. [PMID: 25753324 PMCID: PMC4574834 DOI: 10.1111/bcp.12624] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 02/05/2015] [Accepted: 03/03/2015] [Indexed: 12/21/2022] Open
Abstract
AIMS The authors' aim was to conduct a proof-of-principle study to test whether c-Jun N-terminal kinase (JNK) phosphorylation and Noxa induction occur in peripheral blood chronic lymphocytic leukaemia (CLL) cells in patients receiving a vincristine infusion. METHODS Patients with CLL received 2 mg vincristine by a 5-min intravenous infusion. Blood samples were collected at baseline and up to 6 h after the vincristine infusion, and assayed for JNK activation, Noxa induction and vincristine plasma concentrations. RESULTS Ex vivo treated peripheral CLL cells activated JNK in response to 10-100 nM vincristine in 6 h. Noxa protein expression, while variable, was also observed over this time frame. In CLL patients, vincristine infusion led to rapid (<1 h) JNK phosphorylation in peripheral blood CLL cells which was sustained for at least 4-6 h after the vincristine infusion. Noxa protein expression was not observed in response to vincristine infusion. CONCLUSIONS This study confirmed that vincristine can activate JNK but not induce Noxa in CLL cells in vivo. The results suggest that novel JNK-dependent drug combinations with vincristine warrant further investigation.
Collapse
MESH Headings
- Antineoplastic Agents, Phytogenic/administration & dosage
- Antineoplastic Agents, Phytogenic/pharmacokinetics
- Antineoplastic Agents, Phytogenic/pharmacology
- Enzyme Activation/drug effects
- Humans
- Infusions, Intravenous
- JNK Mitogen-Activated Protein Kinases/blood
- JNK Mitogen-Activated Protein Kinases/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/blood
- Leukemia, Lymphocytic, Chronic, B-Cell/enzymology
- Lymphocytes/drug effects
- Lymphocytes/enzymology
- Phosphorylation
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Vincristine/administration & dosage
- Vincristine/pharmacokinetics
- Vincristine/pharmacology
Collapse
Affiliation(s)
- Darcy J P Bates
- Department of MedicineLebanon, NH, 03756, USA
- Norris Cotton Cancer Center; Geisel School of Medicine at Dartmouth, One Medical Center DriveLebanon, NH, 03756, USA
| | - Lionel D Lewis
- Department of MedicineLebanon, NH, 03756, USA
- Department of Pharmacology and ToxicologyLebanon, NH, 03756, USA
- Norris Cotton Cancer Center; Geisel School of Medicine at Dartmouth, One Medical Center DriveLebanon, NH, 03756, USA
| | - Alan Eastman
- Department of Pharmacology and ToxicologyLebanon, NH, 03756, USA
- Norris Cotton Cancer Center; Geisel School of Medicine at Dartmouth, One Medical Center DriveLebanon, NH, 03756, USA
| | - Alexey V Danilov
- Department of MedicineLebanon, NH, 03756, USA
- Norris Cotton Cancer Center; Geisel School of Medicine at Dartmouth, One Medical Center DriveLebanon, NH, 03756, USA
| |
Collapse
|
22
|
Tawil M, Bekdash A, Mroueh M, Daher CF, Abi-Habib RJ. Wild Carrot Oil Extract is Selectively Cytotoxic to Human Acute Myeloid Leukemia Cells. Asian Pac J Cancer Prev 2015; 16:761-7. [DOI: 10.7314/apjcp.2015.16.2.761] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
23
|
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
|
24
|
Albert MC, Brinkmann K, Kashkar H. Noxa and cancer therapy: Tuning up the mitochondrial death machinery in response to chemotherapy. Mol Cell Oncol 2014; 1:e29906. [PMID: 27308315 PMCID: PMC4905168 DOI: 10.4161/mco.29906] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 06/24/2014] [Accepted: 07/02/2014] [Indexed: 12/12/2022]
Abstract
Biochemical analyses have characterized the BH3-only protein family member Noxa as a “sensitizer” with weak pro-apoptotic activity. Investigations into cancer cell responses to chemotherapeutic agents have identified Noxa as a pivotal factor mediating the cytotoxic effect of a plethora of anticancer treatments independent of its own pro-apoptotic activity. Accumulating evidence now suggests that tumor cells exert a number of strategies to counteract Noxa function by exploiting diverse cellular regulatory circuits that normally govern Noxa expression during cellular stress responses. Here, we summarize data concerning the role of Noxa in cancer chemosensitivity and highlight the potential of this enigmatic BH3-only protein family member in current and novel anticancer therapies.
Collapse
Affiliation(s)
- Marie-Christine Albert
- Centre for Molecular Medicine Cologne (CMMC); Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD); University of Cologne, Köln, Germany; Institute for Medical Microbiology, Immunology and Hygiene (IMMIH); University of Cologne, Köln, Germany
| | - Kerstin Brinkmann
- Centre for Molecular Medicine Cologne (CMMC); Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD); University of Cologne, Köln, Germany; Institute for Medical Microbiology, Immunology and Hygiene (IMMIH); University of Cologne, Köln, Germany
| | - Hamid Kashkar
- Centre for Molecular Medicine Cologne (CMMC); Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD); University of Cologne, Köln, Germany; Institute for Medical Microbiology, Immunology and Hygiene (IMMIH); University of Cologne, Köln, Germany
| |
Collapse
|
25
|
Danilov AV, Soderquist RS, Bates DJP, Eastman A. Toward a cure for chronic lymphocytic leukemia: an attack on multiple fronts. Expert Rev Anticancer Ther 2014; 13:1009-12. [PMID: 24053199 DOI: 10.1586/14737140.2013.825424] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Alexey V Danilov
- Medicine, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | | | | | | |
Collapse
|
26
|
Wang W, Wang YQ, Meng T, Yi JM, Huan XJ, Ma LP, Tong LJ, Chen Y, Ding J, Shen JK, Miao ZH. MCL-1 degradation mediated by JNK activation via MEKK1/TAK1-MKK4 contributes to anticancer activity of new tubulin inhibitor MT189. Mol Cancer Ther 2014; 13:1480-91. [PMID: 24688049 DOI: 10.1158/1535-7163.mct-13-0629] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Colchicine site-targeted tubulin inhibitors are a promising type of anticancer drugs. MT189 is a new derivative of MT119, a previously reported colchicine site-binding antitubulin agent. In this study, MT189 was demonstrated to retain the property of MT119 in disrupting microtubulin via binding to the colchicine site, causing mitotic arrest and inducing apoptosis, and to display 8.7-fold enhanced proliferative inhibition in a panel of cancer cells. MT189 was shown to elicit in vivo anticancer effects on MDA-MB-231 xenografts in nude mice, and the tumor growth was suppressed by 35.9% over 14 days. MT189 led to degradation of MCL-1, a member of the antiapoptotic BCL-2 protein family. Its overexpression reduced but its silenced expression increased the apoptotic induction followed by the treatment with MT189. Moreover, the treatment with MT189 caused activation of the MEKK1/TAK1-MKK4-JNK signaling pathway. The activated JNK resulted in phosphorylation of MCL-1, which facilitated its ubiquitination-mediated degradation. Our results show that MT189 inhibits microtubulin polymerization by binding to the colchicine site. Relief of apoptotic suppression by MCL-1 degradation together with mitotic arrest contributes to the anticancer activity of MT189.
Collapse
Affiliation(s)
- Wei Wang
- Authors' Affiliations: Division of Antitumor Pharmacology and Division of Medicinal Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Ying-Qing Wang
- Authors' Affiliations: Division of Antitumor Pharmacology and Division of Medicinal Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Tao Meng
- Authors' Affiliations: Division of Antitumor Pharmacology and Division of Medicinal Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Jun-Mei Yi
- Authors' Affiliations: Division of Antitumor Pharmacology and Division of Medicinal Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Xia-Juan Huan
- Authors' Affiliations: Division of Antitumor Pharmacology and Division of Medicinal Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Lan-Ping Ma
- Authors' Affiliations: Division of Antitumor Pharmacology and Division of Medicinal Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Lin-Jiang Tong
- Authors' Affiliations: Division of Antitumor Pharmacology and Division of Medicinal Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Yi Chen
- Authors' Affiliations: Division of Antitumor Pharmacology and Division of Medicinal Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Jian Ding
- Authors' Affiliations: Division of Antitumor Pharmacology and Division of Medicinal Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Jing-Kang Shen
- Authors' Affiliations: Division of Antitumor Pharmacology and Division of Medicinal Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Ze-Hong Miao
- Authors' Affiliations: Division of Antitumor Pharmacology and Division of Medicinal Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| |
Collapse
|