1
|
Zhu XQ, Yang H, Lin MH, Shang HX, Peng J, Chen WJ, Chen XZ, Lin JM. Qingjie Fuzheng Granules regulates cancer cell proliferation, apoptosis and tumor angiogenesis in colorectal cancer xenograft mice via Sonic Hedgehog pathway. J Gastrointest Oncol 2021; 11:1123-1134. [PMID: 33456987 DOI: 10.21037/jgo-20-213] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background Sonic Hedgehog (SHh) signaling pathway plays a critical role in cell proliferation, apoptosis, and tumor angiogenesis in various types of malignancies including colorectal cancer (CRC). Qingjie Fuzheng Granules (QFG) is a traditional Chinese medicinal formula, which has been clinically used in various cancer treatments, including CRC. In this study, we explored the potential molecular mechanisms of QFG treatment effects on CRC via the SHh pathway. Methods A CRC HCT-116 xenograft mouse model was utilized for all experiments. Mice were treated with intra-gastric administration of 1 g/kg of QFG or saline 6 days a week for 28 days (4 weeks). Body weight, length and shortest diameter of the tumor were measured every 3 days. At the end of the treatment, the tumor weight was measured. TUNEL staining assays were used to detect tumor apoptosis. Western blot and immunohistochemistry (IHC) assays were used to detect the expression of relative proteins. Results In our results, QFG inhibited the increase of tumor volume and weight, and exhibited no impact on mouse body weight. Furthermore, QFG significantly decreased the expression of SHh, Smo and Gli proteins, indicating the action of SHh signaling. Consequently, the expression of pro-proliferative survivin, Ki-67, Cyclin-D1 and CDK4 were decreased and expression of anti-proliferative p21 was increased. The pro-apoptotic Bax/Bcl-2 ratio, cle-caspase-3 and TUNEL-positive cell percentage in tumor tissues were increased. Meanwhile, the pro-angiogenic VEGF-A and VEGFR-2 expression was down-regulated. Conclusions QFG inhibited CRC cell proliferation and promoted CRC cell apoptosis and tumor angiogenesis in vivo through the suppression of SHh pathway, suggesting that QFG could be a potential therapeutic drug for CRC.
Collapse
Affiliation(s)
- Xiao-Qin Zhu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, China
| | - Hong Yang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, China
| | - Ming-He Lin
- Editorial Department of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Hai-Xia Shang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, China
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, China
| | - Wu-Jin Chen
- Department of Oncology, Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xu-Zheng Chen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, China
| | - Jiu-Mao Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, China
| |
Collapse
|
2
|
Wilde L, Ramanathan S, Kasner M. B-cell lymphoma-2 inhibition and resistance in acute myeloid leukemia. World J Clin Oncol 2020; 11:528-540. [PMID: 32879842 PMCID: PMC7443828 DOI: 10.5306/wjco.v11.i8.528] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 07/01/2020] [Accepted: 07/19/2020] [Indexed: 02/06/2023] Open
Abstract
Spurred by better understanding of disease biology, improvements in molecular diagnostics, and the development of targeted therapies, the treatment of acute myeloid leukemia (AML) has undergone significant evolution in recent years. Arguably, the most exciting shift has come from the success of treatment with the B-cell lymphoma-2 inhibitor venetoclax. When given in combination with a hypomethylating agent or low dose cytarabine, venetoclax demonstrates high response rates, some of which are durable. In spite of this, relapses after venetoclax treatment are common, and much interest exists in elucidating the mechanisms of resistance to the drug. Alterations in leukemic stem cell metabolism have been identified as a possible escape route, and clinical trials focusing on targeting metabolism in AML are ongoing. This review article highlights current research regarding venetoclax treatment and resistance in AML with a focus on cellular metabolism.
Collapse
Affiliation(s)
- Lindsay Wilde
- Department of Hematology and Medical Oncology, Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital, Philadelphia, PA 19107, United States
| | - Sabarina Ramanathan
- Department of Hematology and Medical Oncology, Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital, Philadelphia, PA 19107, United States
| | - Margaret Kasner
- Department of Hematology and Medical Oncology, Sidney Kimmel Cancer Center at Thomas Jefferson University Hospital, Philadelphia, PA 19107, United States
| |
Collapse
|
3
|
Aspartic Aminopeptidase Is a Novel Biomarker of Aggressive Chronic Lymphocytic Leukemia. Cancers (Basel) 2020; 12:cancers12071876. [PMID: 32664705 PMCID: PMC7408864 DOI: 10.3390/cancers12071876] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 11/17/2022] Open
Abstract
Treatment of chronic lymphocytic leukemia has advanced substantially as our understanding of the kinase signal transduction pathways driven by the B cell receptor (BcR) has developed. Particularly, understanding the role of Bruton tyrosine kinase and phosphatidyl inositol 3 kinase delta in driving prosurvival signal transduction in chronic lymphocytic leukemia (CLL) cells and their targeting with pharmacological inhibitors (ibrutinib and idelalisib, respectively) has improved patient outcomes significantly. The kinase signaling pathway induced by the BcR is highly complex and has multiple interconnecting branches mediated by tyrosine and serine/threonine kinases activated downstream of the BcR. There is a high level of redundancy in the biological responses, with several BcR-signaling kinases driving nuclear factor kappa B activation or inducing antiapoptotic Bcl-2 genes. Accordingly, common gene targets of BcR-signaling kinases may serve as biomarkers indicating enhanced BCR-signaling and aggressive disease progression. This study used a gene expression correlation analysis of malignant B cell lines and primary CLL cells to identify genes whose expression correlated with BCR-signaling kinases overexpressed and/or overactivated in CLL, namely: AKT1, AKT2, BTK, MAPK1, MAPK3, PI3KCD and ZAP70. The analysis identified a 32-gene signature with a strong prognostic potential and DNPEP, the gene coding for aspartic aminopeptidase, as a predictor of aggressive CLL. DNPEP gene expression correlated with MAPK3, PI3KCD, and ZAP70 expression and, in the primary CLL test dataset, showed a strong prognostic potential. The inhibition of DNPEP with a pharmacological inhibitor enhanced the cytotoxic potential of idelalisib and ibrutinib, indicating a biological functionality of DNPEP in CLL. DNPEP, as an aminopeptidase, contributes to the maintenance of the free amino acid pool in CLL cells found to be an essential process for the survival of many cancer cell types, and thus, these results warrant further research into the exploitation of aminopeptidase inhibitors in the treatment of drug-resistant CLL.
Collapse
|
4
|
Dhami SPS, Tirincsi A, Baev D, Krawczyk J, Quinn J, Cahill MR, Zeugolis D, Szegezdi E. Theranostic drug test incorporating the bone-marrow microenvironment can predict the clinical response of acute myeloid leukaemia to chemotherapy. Br J Haematol 2020; 189:e254-e258. [PMID: 32342487 DOI: 10.1111/bjh.16684] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Sukhraj Pal S Dhami
- Apoptosis Research Centre, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Andrea Tirincsi
- Apoptosis Research Centre, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Denis Baev
- Apoptosis Research Centre, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Janusz Krawczyk
- Department of Haematology, Galway University Hospital, Galway, Ireland
| | - John Quinn
- Department of Haematology, Beaumont Hospital and Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Mary R Cahill
- Department of Haematology, Cork University Hospital, Cork, Ireland
| | - Dimitrios Zeugolis
- Regenerative, Modular and Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland, Galway, Ireland.,Science Foundation of Ireland, Centre for Research in Medical Devices (CURAM), Biomedical Sciences Building, National University of Ireland, Galway, Ireland
| | - Eva Szegezdi
- Apoptosis Research Centre, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
5
|
Repression of Mcl-1 expression by the CDC7/CDK9 inhibitor PHA-767491 overcomes bone marrow stroma-mediated drug resistance in AML. Sci Rep 2018; 8:15752. [PMID: 30361682 PMCID: PMC6202320 DOI: 10.1038/s41598-018-33982-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 10/03/2018] [Indexed: 02/08/2023] Open
Abstract
Acute myeloid leukaemia (AML) is an aggressive cancer with 50-75% of patients relapsing even after successful chemotherapy. The role of the bone marrow microenvironment (BMM) in protecting AML cells from chemotherapeutics and causing consequent relapse is increasingly recognised. However the role that the anti-apoptotic Bcl-2 proteins play as effectors of BMM-mediated drug resistance are less understood. Here we show that bone marrow mesenchymal stromal cells (BMSC) provide resistance to AML cells against BH3-mimetics, cytarabine and daunorubicin, but this is not mediated by Bcl-2 and/or Bcl-XL as previously thought. Instead, BMSCs induced Mcl-1 expression over Bcl-2 and/or Bcl-XL in AML cells and inhibition of Mcl-1 with a small-molecule inhibitor, A1210477, or repressing its expression with the CDC7/CDK9 dual-inhibitor, PHA-767491 restored sensitivity to BH3-mimetics. Furthermore, combined inhibition of Bcl-2/Bcl-XL and Mcl-1 could revert BMSC-mediated resistance against cytarabine + daunorubicin. Importantly, the CD34+/CD38- leukemic stem cell-encompassing population was equally sensitive to the combination of PHA-767491 and ABT-737. These results indicate that Bcl-2/Bcl-XL and Mcl-1 act in a redundant fashion as effectors of BMM-mediated AML drug resistance and highlight the potential of Mcl-1-repression to revert BMM-mediated drug resistance in the leukemic stem cell population, thus, prevent disease relapse and ultimately improve patient survival.
Collapse
|
6
|
Shutting Down Acute Myeloid Leukemia and Myelodysplastic Syndrome with BCL-2 Family Protein Inhibition. Curr Hematol Malig Rep 2018; 13:256-264. [DOI: 10.1007/s11899-018-0464-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
7
|
Anderson GR, Wardell SE, Cakir M, Crawford L, Leeds JC, Nussbaum DP, Shankar PS, Soderquist RS, Stein EM, Tingley JP, Winter PS, Zieser-Misenheimer EK, Alley HM, Yllanes A, Haney V, Blackwell KL, McCall SJ, McDonnell DP, Wood KC. PIK3CA mutations enable targeting of a breast tumor dependency through mTOR-mediated MCL-1 translation. Sci Transl Med 2017; 8:369ra175. [PMID: 27974663 DOI: 10.1126/scitranslmed.aae0348] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/06/2016] [Accepted: 10/05/2016] [Indexed: 12/23/2022]
Abstract
Therapies that efficiently induce apoptosis are likely to be required for durable clinical responses in patients with solid tumors. Using a pharmacological screening approach, we discovered that combined inhibition of B cell lymphoma-extra large (BCL-XL) and the mammalian target of rapamycin (mTOR)/4E-BP axis results in selective and synergistic induction of apoptosis in cellular and animal models of PIK3CA mutant breast cancers, including triple-negative tumors. Mechanistically, inhibition of mTOR/4E-BP suppresses myeloid cell leukemia-1 (MCL-1) protein translation only in PIK3CA mutant tumors, creating a synthetic dependence on BCL-XL This dual dependence on BCL-XL and MCL-1, but not on BCL-2, appears to be a fundamental property of diverse breast cancer cell lines, xenografts, and patient-derived tumors that is independent of the molecular subtype or PIK3CA mutational status. Furthermore, this dependence distinguishes breast cancers from normal breast epithelial cells, which are neither primed for apoptosis nor dependent on BCL-XL/MCL-1, suggesting a potential therapeutic window. By tilting the balance of pro- to antiapoptotic signals in the mitochondria, dual inhibition of MCL-1 and BCL-XL also sensitizes breast cancer cells to standard-of-care cytotoxic and targeted chemotherapies. Together, these results suggest that patients with PIK3CA mutant breast cancers may benefit from combined treatment with inhibitors of BCL-XL and the mTOR/4E-BP axis, whereas alternative methods of inhibiting MCL-1 and BCL-XL may be effective in tumors lacking PIK3CA mutations.
Collapse
Affiliation(s)
- Grace R Anderson
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Suzanne E Wardell
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Merve Cakir
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA.,Program in Computational Biology and Bioinformatics, Duke University, Durham, NC 27708, USA
| | - Lorin Crawford
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA.,Department of Statistical Science, Duke University, Durham, NC 27708, USA
| | - Jim C Leeds
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Daniel P Nussbaum
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA.,Department of Surgery, Duke University, Durham, NC 27710, USA
| | - Pallavi S Shankar
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Ryan S Soderquist
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Elizabeth M Stein
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Jennifer P Tingley
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Peter S Winter
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA.,Program in Genetics and Genomics, Duke University, Durham, NC 27710, USA
| | | | - Holly M Alley
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Alexander Yllanes
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Victoria Haney
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | | | | | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Kris C Wood
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
8
|
Gilormini M, Malesys C, Armandy E, Manas P, Guy JB, Magné N, Rodriguez-Lafrasse C, Ardail D. Preferential targeting of cancer stem cells in the radiosensitizing effect of ABT-737 on HNSCC. Oncotarget 2017; 7:16731-44. [PMID: 26934442 PMCID: PMC4941347 DOI: 10.18632/oncotarget.7744] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/13/2016] [Indexed: 12/26/2022] Open
Abstract
Head and neck squamous cell carcinomas (HNSCC) are common human malignancies with poor clinical outcomes. The 5-year survival rates for patients with advanced stage HNSCC have not changed appreciably in the past few decades, underscoring a dire need for improved therapeutic options. HNSCC is frequently characterized by overexpression of anti-apoptotic Bcl-2 family members. Increased levels of these anti-apoptotic proteins have been associated with radio- and chemoresistance and poor clinical outcome. The aim of this study was to evaluate combined effects of radiation and ABT-737, a BH3-mimetic molecule, in HNSCC. Although ABT-737, as a single agent, was largely ineffective at promoting HNSCC cell death, we found that combining ABT-737 and radiation induced strong synergistic apoptosis in HNSCC cell lines and delayed tumoral growth in vivo. Moreover, we demonstrated for the first time that ABT-737, alone or in combination with radiation, can efficiently eliminate cancer stem cells (CSCs). Altogether, our results indicate that therapy targeting anti-apoptotic Bcl-2 family members could be a highly effective potential adjuvant to radiotherapy capable of targeting CSCs in HNSCC and therefore overcoming cancer recurrence and metastasis.
Collapse
Affiliation(s)
- Marion Gilormini
- Université Lyon I, Faculté de Médecine-Lyon-Sud, Oullins, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, EMR3738, Oullins, France
| | - Céline Malesys
- Université Lyon I, Faculté de Médecine-Lyon-Sud, Oullins, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, EMR3738, Oullins, France
| | - Emma Armandy
- Université Lyon I, Faculté de Médecine-Lyon-Sud, Oullins, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, EMR3738, Oullins, France
| | - Patrick Manas
- UMS3444 BioSciences Gerland-Lyon Sud, PBES, Lyon, France
| | - Jean-Baptiste Guy
- Université Lyon I, Faculté de Médecine-Lyon-Sud, Oullins, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, EMR3738, Oullins, France
| | - Nicolas Magné
- Université Lyon I, Faculté de Médecine-Lyon-Sud, Oullins, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, EMR3738, Oullins, France.,Institut de Cancérologie L. Neuwirth, St Etienne, France
| | - Claire Rodriguez-Lafrasse
- Université Lyon I, Faculté de Médecine-Lyon-Sud, Oullins, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, EMR3738, Oullins, France.,Hospices-Civils-de-Lyon, CHLS, Pierre-Bénite, France
| | - Dominique Ardail
- Université Lyon I, Faculté de Médecine-Lyon-Sud, Oullins, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, EMR3738, Oullins, France.,Hospices-Civils-de-Lyon, CHLS, Pierre-Bénite, France
| |
Collapse
|
9
|
Minagawa K, Jamil MO, AL-Obaidi M, Pereboeva L, Salzman D, Erba HP, Lamb LS, Bhatia R, Mineishi S, Di Stasi A. In Vitro Pre-Clinical Validation of Suicide Gene Modified Anti-CD33 Redirected Chimeric Antigen Receptor T-Cells for Acute Myeloid Leukemia. PLoS One 2016; 11:e0166891. [PMID: 27907031 PMCID: PMC5132227 DOI: 10.1371/journal.pone.0166891] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 11/04/2016] [Indexed: 12/20/2022] Open
Abstract
Background Approximately fifty percent of patients with acute myeloid leukemia can be cured with current therapeutic strategies which include, standard dose chemotherapy for patients at standard risk of relapse as assessed by cytogenetic and molecular analysis, or high-dose chemotherapy with allogeneic hematopoietic stem cell transplant for high-risk patients. Despite allogeneic hematopoietic stem cell transplant about 25% of patients still succumb to disease relapse, therefore, novel strategies are needed to improve the outcome of patients with acute myeloid leukemia. Methods and findings We developed an immunotherapeutic strategy targeting the CD33 myeloid antigen, expressed in ~ 85–90% of patients with acute myeloid leukemia, using chimeric antigen receptor redirected T-cells. Considering that administration of CAR T-cells has been associated with cytokine release syndrome and other potential off-tumor effects in patients, safety measures were here investigated and reported. We genetically modified human activated T-cells from healthy donors or patients with acute myeloid leukemia with retroviral supernatant encoding the inducible Caspase9 suicide gene, a ΔCD19 selectable marker, and a humanized third generation chimeric antigen receptor recognizing human CD33. ΔCD19 selected inducible Caspase9-CAR.CD33 T-cells had a 75±3.8% (average ± standard error of the mean) chimeric antigen receptor expression, were able to specifically lyse CD33+ targets in vitro, including freshly isolated leukemic blasts from patients, produce significant amount of tumor-necrosis-factor-alpha and interferon-gamma, express the CD107a degranulation marker, and proliferate upon antigen specific stimulation. Challenging ΔCD19 selected inducible Caspase9-CAR.CD33 T-cells with programmed-death-ligand-1 enriched leukemia blasts resulted in significant killing like observed for the programmed-death-ligand-1 negative leukemic blasts fraction. Since the administration of 10 nanomolar of a non-therapeutic dimerizer to activate the suicide gene resulted in the elimination of only 76.4±2.0% gene modified cells in vitro, we found that co-administration of the dimerizer with either the BCL-2 inhibitor ABT-199, the pan-BCL inhibitor ABT-737, or mafosfamide, resulted in an additive effect up to complete cell elimination. Conclusions This strategy could be investigated for the safety of CAR T-cell applications, and targeting CD33 could be used as a ‘bridge” therapy for patients coming to allogeneic hematopoietic stem cell transplant, as anti-leukemia activity from infusing CAR.CD33 T-cells has been demonstrated in an ongoing clinical trial. Albeit never performed in the clinical setting, our future plan is to investigate the utility of iC9-CAR.CD33 T-cells as part of the conditioning therapy for an allogeneic hematopoietic stem cell transplant for acute myeloid leukemia, together with other myelosuppressive agents, whilst the activation of the inducible Caspase9 suicide gene would grant elimination of the infused gene modified T-cells prior to stem cell infusion to reduce the risk of engraftment failure as the CD33 is also expressed on a proportion of the donor stem cell graft.
Collapse
MESH Headings
- B7-H1 Antigen/pharmacology
- Biphenyl Compounds/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Caspase 9/genetics
- Caspase 9/immunology
- Cell Engineering
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cellular Reprogramming
- Clinical Trials as Topic
- Cyclophosphamide/analogs & derivatives
- Cyclophosphamide/pharmacology
- Cytotoxicity, Immunologic
- Genetic Vectors
- Humans
- Interferon-gamma/biosynthesis
- Interferon-gamma/immunology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Lysosomal-Associated Membrane Protein 1/genetics
- Lysosomal-Associated Membrane Protein 1/immunology
- Myeloid Cells/drug effects
- Myeloid Cells/immunology
- Myeloid Cells/pathology
- Nitrophenols/pharmacology
- Piperazines/pharmacology
- Primary Cell Culture
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors
- Sialic Acid Binding Ig-like Lectin 3/genetics
- Sialic Acid Binding Ig-like Lectin 3/immunology
- Sulfonamides/pharmacology
- T-Lymphocytes/cytology
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- Tumor Necrosis Factor-alpha/biosynthesis
- Tumor Necrosis Factor-alpha/immunology
Collapse
Affiliation(s)
- Kentaro Minagawa
- Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Muhammad O. Jamil
- Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Mustafa AL-Obaidi
- Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Larisa Pereboeva
- Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Donna Salzman
- Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Harry P. Erba
- Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Lawrence S. Lamb
- Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Ravi Bhatia
- Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Shin Mineishi
- Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Antonio Di Stasi
- Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
- * E-mail:
| |
Collapse
|
10
|
Wang T, Shigdar S, Gantier MP, Hou Y, Wang L, Li Y, Shamaileh HA, Yin W, Zhou SF, Zhao X, Duan W. Cancer stem cell targeted therapy: progress amid controversies. Oncotarget 2016; 6:44191-206. [PMID: 26496035 PMCID: PMC4792551 DOI: 10.18632/oncotarget.6176] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 10/06/2015] [Indexed: 12/12/2022] Open
Abstract
Although cancer stem cells have been well characterized in numerous malignancies, the fundamental characteristics of this group of cells, however, have been challenged by some recent observations: cancer stem cells may not necessary to be rare within tumors; cancer stem cells and non-cancer stem cells may undergo reversible phenotypic changes; and the cancer stem cells phenotype can vary substantially between patients. Here the current status and progresses of cancer stem cells theory is illustrated and via providing a panoramic view of cancer therapy, we addressed the recent controversies regarding the feasibility of cancer stem cells targeted anti-cancer therapy.
Collapse
Affiliation(s)
- Tao Wang
- School of Nursing, Zhengzhou University, Zhengzhou, China.,School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Sarah Shigdar
- School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Michael P Gantier
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Yingchun Hou
- Co-Innovation Center for Qinba Region's Sustainable Development, Shaanxi Normal University, Xi'an, China
| | - Li Wang
- Department of Gynecologic Oncology, Henan Cancer Hospital, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Yong Li
- Cancer Care Centre, St George Hospital and St George Clinical School, University of New South Wales (UNSW), Kensington, Australia
| | - Hadi Al Shamaileh
- School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Wang Yin
- School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Shu-Feng Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Xinhan Zhao
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Wei Duan
- School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| |
Collapse
|
11
|
Inhibition of Bcl-2 family members sensitises soft tissue leiomyosarcomas to chemotherapy. Br J Cancer 2016; 114:1219-26. [PMID: 27140314 PMCID: PMC4891502 DOI: 10.1038/bjc.2016.117] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 03/11/2016] [Accepted: 04/07/2016] [Indexed: 12/17/2022] Open
Abstract
Background: Leiomyosarcoma is an aggressive soft tissue sarcoma with a 5-year survival rate of 15 to 60%. Treatment options for inoperable or metastatic patients are limited owing to frequent resistance of tumours to chemotherapy and radiation. In this study, we hypothesised that antiapoptotic Bcl-2 family proteins might contribute to leiomyosarcoma chemoresistance and therefore inhibition of Bcl-2 family proteins might sensitise leiomyosarcomas to conventional chemotherapy. Methods: Expression of the Bcl-2 family proteins Bcl-xL, Bcl-w and Bcl-2 was investigated using immunohistochemistry on a tissue microarray containing 43 leiomyosarcomas. Furthermore, we investigated whether ABT-737, a potent BH3 mimetic, sensitises leiomyosarcoma cells to doxorubicin treatment in vitro. Results: Seventy-seven per cent, 84% and 42% of leiomyosarcomas demonstrated high expression of Bcl-2, Bcl-xL and Bcl-w, respectively. Single-agent treatment with ABT-737 resulted in a minor reduction of cell viability. However, combination treatment of ABT-737 and doxorubicin revealed synergism in all four cell lines, by inducing apoptosis. Conclusions: In conclusion, Bcl-2 family proteins contribute to soft tissue leiomyosarcoma chemoresistance. Antiapoptotic proteins are highly expressed in leiomyosarcoma of soft tissue, and inhibition of these proteins using a BH3 mimetic increases leiomyosarcoma sensitivity to doxorubicin.
Collapse
|