1
|
Li Z, Zou J, Chen X. In Response to Precision Medicine: Current Subcellular Targeting Strategies for Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2209529. [PMID: 36445169 DOI: 10.1002/adma.202209529] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/08/2022] [Indexed: 05/26/2023]
Abstract
Emerging as a potent anticancer treatment, subcellular targeted cancer therapy has drawn increasing attention, bringing great opportunities for clinical application. Here, two targeting strategies for four main subcellular organelles (mitochondria, lysosome, endoplasmic reticulum, and nucleus), including molecule- and nanomaterial (inorganic nanoparticles, micelles, organic polymers, and others)-based targeted delivery or therapeutic strategies, are summarized. Phototherapy, chemotherapy, radiotherapy, immunotherapy, and "all-in-one" combination therapy are among the strategies covered in detail. Such materials are constructed based on the specific properties and relevant mechanisms of organelles, enabling the elimination of tumors by inducing dysfunction in the corresponding organelles or destroying specific structures. The challenges faced by organelle-targeting cancer therapies are also summarized. Looking forward, a paradigm for organelle-targeting therapy with enhanced therapeutic efficacy compared to current clinical approaches is envisioned.
Collapse
Affiliation(s)
- Zheng Li
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Jianhua Zou
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|
2
|
Zhao S, Tang Y, Wang R, Najafi M. Mechanisms of cancer cell death induction by paclitaxel: an updated review. Apoptosis 2022; 27:647-667. [PMID: 35849264 DOI: 10.1007/s10495-022-01750-z] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2022] [Indexed: 02/07/2023]
Abstract
Chemoresistance of cancer cells is a major problem in treating cancer. Knowledge of how cancer cells may die or resist cancer drugs is critical to providing certain strategies to overcome tumour resistance to treatment. Paclitaxel is known as a chemotherapy drug that can suppress the proliferation of cancer cells by inducing cell cycle arrest and induction of mitotic catastrophe. However, today, it is well known that paclitaxel can induce multiple kinds of cell death in cancers. Besides the induction of mitotic catastrophe that occurs during mitosis, paclitaxel has been shown to induce the expression of several pro-apoptosis mediators. It also can modulate the activity of anti-apoptosis mediators. However, certain cell-killing mechanisms such as senescence and autophagy can increase resistance to paclitaxel. This review focuses on the mechanisms of cell death, including apoptosis, mitotic catastrophe, senescence, autophagic cell death, pyroptosis, etc., following paclitaxel treatment. In addition, mechanisms of resistance to cell death due to exposure to paclitaxel and the use of combinations to overcome drug resistance will be discussed.
Collapse
Affiliation(s)
- Shuang Zhao
- School of Basic Medicine, Shaoyang University, Shaoyang, 422000, Hunan, China.
| | - Yufei Tang
- College of Medical Technology, Shaoyang University, Shaoyang, 422000, Hunan, China
| | - Ruohan Wang
- School of Nursing, Shaoyang University, Shaoyang, 422000, Hunan, China.
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
3
|
Karbon G, Haschka MD, Hackl H, Soratroi C, Rocamora-Reverte L, Parson W, Fiegl H, Villunger A. The BH3-only protein NOXA serves as an independent predictor of breast cancer patient survival and defines susceptibility to microtubule targeting agents. Cell Death Dis 2021; 12:1151. [PMID: 34903710 PMCID: PMC8668920 DOI: 10.1038/s41419-021-04415-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 11/11/2021] [Accepted: 11/22/2021] [Indexed: 12/21/2022]
Abstract
Breast cancer (BC) treatment frequently involves microtubule-targeting agents (MTAs), such as paclitaxel, that arrest cells in mitosis. Sensitivity to MTAs is defined by a subset of pro- and anti-apoptotic BCL2 family proteins controlling mitochondrial apoptosis. Here, we aimed to determine their prognostic value in primary tumour samples from 92 BC patients. Our analysis identified high NOXA/PMAIP mRNA expression levels as an independent prognostic marker for improved relapse-free survival (RFS) and overall survival (OS) in multivariate analysis in BC patients, independent of their molecular subtype. Analysis of available TCGA datasets of 1060 BC patients confirmed our results and added a clear predictive value of NOXA mRNA levels for patients who received MTA-based therapy. In this TCGA cohort, 122 patients received MTA-treatment and high NOXA mRNA levels correlated with their progression-free interval (PFI) and OS. Our follow-up analyses in a panel of BC cell lines of different molecular subtypes identified NOXA protein expression as a key determinant of paclitaxel sensitivity in triple-negative breast cancer (TNBC) cells. Moreover, we noted highest additive effects between paclitaxel and chemical inhibition of BCLX, but not BCL2 or MCL1, documenting dependence of TNBC cells on BCLX for survival and paclitaxel sensitivity defined by NOXA expression levels.
Collapse
Affiliation(s)
- Gerlinde Karbon
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Manuel D Haschka
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Hubert Hackl
- Insitute for Bioinformatics, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Claudia Soratroi
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Lourdes Rocamora-Reverte
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Walther Parson
- Institute of Legal Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Heidelinde Fiegl
- Department for Obstetrics & Gynecology, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas Villunger
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
4
|
Astarita EM, Maloney SM, Hoover CA, Berkeley BJ, VanKlompenberg MK, Nair TM, Prosperi JR. Adenomatous Polyposis Coli loss controls cell cycle regulators and response to paclitaxel in MDA-MB-157 metaplastic breast cancer cells. PLoS One 2021; 16:e0255738. [PMID: 34370741 PMCID: PMC8351968 DOI: 10.1371/journal.pone.0255738] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 07/22/2021] [Indexed: 11/22/2022] Open
Abstract
Adenomatous Polyposis Coli (APC) is lost in approximately 70% of sporadic breast cancers, with an inclination towards triple negative breast cancer (TNBC). TNBC is treated with traditional chemotherapy, such as paclitaxel (PTX); however, tumors often develop drug resistance. We previously created APC knockdown cells (APC shRNA1) using the human TNBC cells, MDA-MB-157, and showed that APC loss induces PTX resistance. To understand the mechanisms behind APC-mediated PTX response, we performed cell cycle analysis and analyzed cell cycle related proteins. Cell cycle analysis indicated increased G2/M population in both PTX-treated APC shRNA1 and parental cells, suggesting that APC expression does not alter PTX-induced G2/M arrest. We further studied the subcellular localization of the G2/M transition proteins, cyclin B1 and CDK1. The APC shRNA1 cells had increased CDK1, which was preferentially localized to the cytoplasm, and increased baseline CDK6. RNA-sequencing was performed to gain a global understanding of changes downstream of APC loss and identified a broad mis-regulation of cell cycle-related genes in APC shRNA1 cells. Our studies are the first to show an interaction between APC and taxane response in breast cancer. The implications include designing combination therapy to re-sensitize APC-mutant breast cancers to taxanes using the specific cell cycle alterations.
Collapse
Affiliation(s)
- Emily M. Astarita
- Harper Cancer Research Institute, South Bend, IN, United States of America
- Department of Chemistry/Biochemistry, University of Notre Dame, Notre Dame, IN, United States of America
| | - Sara M. Maloney
- Harper Cancer Research Institute, South Bend, IN, United States of America
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, South Bend, South Bend, IN, United States of America
| | - Camden A. Hoover
- Harper Cancer Research Institute, South Bend, IN, United States of America
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States of America
| | | | - Monica K. VanKlompenberg
- Harper Cancer Research Institute, South Bend, IN, United States of America
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, South Bend, South Bend, IN, United States of America
| | - T. Murlidharan Nair
- Department of Biology and Computer Science/Informatics, Indiana University South Bend, South Bend, IN, United States of America
| | - Jenifer R. Prosperi
- Harper Cancer Research Institute, South Bend, IN, United States of America
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, South Bend, South Bend, IN, United States of America
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States of America
| |
Collapse
|
5
|
Pairawan S, Zhao M, Yuca E, Annis A, Evans K, Sutton D, Carvajal L, Ren JG, Santiago S, Guerlavais V, Akcakanat A, Tapia C, Yang F, Bose PSC, Zheng X, Dumbrava EI, Aivado M, Meric-Bernstam F. First in class dual MDM2/MDMX inhibitor ALRN-6924 enhances antitumor efficacy of chemotherapy in TP53 wild-type hormone receptor-positive breast cancer models. Breast Cancer Res 2021; 23:29. [PMID: 33663585 PMCID: PMC7934277 DOI: 10.1186/s13058-021-01406-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 02/17/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND MDM2/MDMX proteins are frequently elevated in hormone receptor-positive (ER+) breast cancer. We sought to determine the antitumor efficacy of the combination of ALRN-6924, a dual inhibitor of MDM2/MDMX, with chemotherapy in ER+ breast cancer models. METHODS Three hundred two cell lines representing multiple tumor types were screened to confirm the role of TP53 status in ALRN-6924 efficacy. ER+ breast cancer cell lines (MCF-7 and ZR-75-1) were used to investigate the antitumor efficacy of ALRN-6924 combination. In vitro cell proliferation, cell cycle, and apoptosis assays were performed. Xenograft tumor volumes were measured, and reverse-phase protein array (RPPA), immunohistochemistry (IHC), and TUNEL assay of tumor tissues were performed to evaluate the in vivo pharmacodynamic effects of ALRN-6924 with paclitaxel. RESULTS ALRN-6924 was active in wild-type TP53 (WT-TP53) cancer cell lines, but not mutant TP53. On ER+ breast cancer cell lines, it was synergistic in vitro and had enhanced in vivo antitumor activity with both paclitaxel and eribulin. Flow cytometry revealed signs of mitotic crisis in all treatment groups; however, S phase was only decreased in MCF-7 single agent and combinatorial ALRN-6924 arms. RPPA and IHC demonstrated an increase in p21 expression in both combinatorial and single agent ALRN-6924 in vivo treatment groups. Apoptotic assays revealed a significantly enhanced in vivo apoptotic rate in ALRN-6924 combined with paclitaxel treatment arm compared to either single agent. CONCLUSION The significant synergy observed with ALRN-6924 in combination with chemotherapeutic agents supports further evaluation in patients with hormone receptor-positive breast cancer.
Collapse
Affiliation(s)
- Seyed Pairawan
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ming Zhao
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Blvd, Houston, TX, 77030, USA
| | - Erkan Yuca
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Blvd, Houston, TX, 77030, USA
| | | | - Kurt Evans
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Blvd, Houston, TX, 77030, USA
| | | | | | | | | | | | - Argun Akcakanat
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Blvd, Houston, TX, 77030, USA
| | - Coya Tapia
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Present address: Epizyme Inc., Cambridge, MA, USA
| | - Fei Yang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Priya Subash Chandra Bose
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaofeng Zheng
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ecaterina Ileana Dumbrava
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Blvd, Houston, TX, 77030, USA
| | | | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Blvd, Houston, TX, 77030, USA.
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
6
|
Čermák V, Dostál V, Jelínek M, Libusová L, Kovář J, Rösel D, Brábek J. Microtubule-targeting agents and their impact on cancer treatment. Eur J Cell Biol 2020; 99:151075. [PMID: 32414588 DOI: 10.1016/j.ejcb.2020.151075] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/25/2020] [Accepted: 03/17/2020] [Indexed: 02/07/2023] Open
Abstract
Microtubule-targeting agents (MTAs) constitute a diverse group of chemical compounds that bind to microtubules and affect their properties and function. Disruption of microtubules induces various cellular responses often leading to cell cycle arrest or cell death, the most common effect of MTAs. MTAs have found a plethora of practical applications in weed control, as fungicides and antiparasitics, and particularly in cancer treatment. Here we summarize the current knowledge of MTAs, the mechanisms of action and their role in cancer treatment. We further outline the potential use of MTAs in anti-metastatic therapy based on inhibition of cancer cell migration and invasiveness. The two main problems associated with cancer therapy by MTAs are high systemic toxicity and development of resistance. Toxic side effects of MTAs can be, at least partly, eliminated by conjugation of the drugs with various carriers. Moreover, some of the novel MTAs overcome the resistance mediated by both multidrug resistance transporters as well as overexpression of specific β-tubulin types. In anti-metastatic therapy, MTAs should be combined with other drugs to target all modes of cancer cell invasion.
Collapse
Affiliation(s)
- Vladimír Čermák
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Vojtěch Dostál
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic
| | - Michael Jelínek
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism, and Nutrition, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Lenka Libusová
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic
| | - Jan Kovář
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism, and Nutrition, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Daniel Rösel
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic.
| |
Collapse
|
7
|
Dubois F, Keller M, Hoflack J, Maille E, Antoine M, Westeel V, Bergot E, Quoix E, Lavolé A, Bigay-Game L, Pujol JL, Langlais A, Morin F, Zalcman G, Levallet G. Role of the YAP-1 Transcriptional Target cIAP2 in the Differential Susceptibility to Chemotherapy of Non-Small-Cell Lung Cancer (NSCLC) Patients with Tumor RASSF1A Gene Methylation from the Phase 3 IFCT-0002 Trial. Cancers (Basel) 2019; 11:cancers11121835. [PMID: 31766357 PMCID: PMC6966477 DOI: 10.3390/cancers11121835] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/12/2019] [Accepted: 11/19/2019] [Indexed: 12/27/2022] Open
Abstract
RASSF1 gene methylation predicts longer disease-free survival (DFS) and overall survival (OS) in patients with early-stage non-small-cell lung cancer treated using paclitaxel-based neo-adjuvant chemotherapy compared to patients receiving a gemcitabine-based regimen, according to the randomized Phase 3 IFCT (Intergroupe Francophone de Cancérologie Thoracique)-0002 trial. To better understand these results, this study used four human bronchial epithelial cell (HBEC) models (HBEC-3, HBEC-3-RasV12, A549, and H1299) and modulated the expression of RASSF1A or YAP-1. Wound-healing, invasion, proliferation and apoptosis assays were then carried out and the expression of YAP-1 transcriptional targets was quantified using a quantitative polymerase chain reaction. This study reports herein that gemcitabine synergizes with RASSF1A, silencing to increase the IAP-2 expression, which in turn not only interferes with cell proliferation but also promotes cell migration. This contributes to the aggressive behavior of RASSF1A-depleted cells, as confirmed by a combined knockdown of IAP-2 and RASSF1A. Conversely, paclitaxel does not increase the IAP-2 expression but limits the invasiveness of RASSF1A-depleted cells, presumably by rescuing microtubule stabilization. Overall, these data provide a functional insight that supports the prognostic value of RASSF1 gene methylation on survival of early-stage lung cancer patients receiving perioperative paclitaxel-based treatment compared to gemcitabine-based treatment, identifying IAP-2 as a novel biomarker indicative of YAP-1-mediated modulation of chemo-sensitivity in lung cancer.
Collapse
Affiliation(s)
- Fatéméh Dubois
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP CYCERON, 14074 Caen, France; (F.D.); (M.K.); (E.M.); (E.B.)
- Department of Pathology, CHU de Caen, 14033 Caen, France
| | - Maureen Keller
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP CYCERON, 14074 Caen, France; (F.D.); (M.K.); (E.M.); (E.B.)
- Normandie Université, UNICAEN, UPRES-EA2608, 14032 Caen, France
| | - Julien Hoflack
- Normandie Université, UNICAEN, UPRES-EA2608, 14032 Caen, France
| | - Elodie Maille
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP CYCERON, 14074 Caen, France; (F.D.); (M.K.); (E.M.); (E.B.)
- Normandie Université, UNICAEN, INSERM UMR 1086 ANTICIPE, 14032 Caen, France
| | - Martine Antoine
- Department of Pathology, Hôpital Tenon, AP-HP, 75020 Paris, France;
| | - Virginie Westeel
- Department of Pneumology, University Hospital of Besançon, University Bourgogne Franche-Comté, 25000 Besançon, France;
| | - Emmanuel Bergot
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP CYCERON, 14074 Caen, France; (F.D.); (M.K.); (E.M.); (E.B.)
- Department of Pulmonology & Thoracic Oncology, CHU de Caen, 14033 Caen, France
| | - Elisabeth Quoix
- Department of Pneumology, University Hospital, 67000 Strasbourg, France;
| | - Armelle Lavolé
- Sorbonne Université, GRC n 04, Theranoscan, AP-HP, Service de Pneumologie, Hôpital Tenon, 75020 Paris, France;
| | - Laurence Bigay-Game
- Pneumology Department, Toulouse-Purpan, University Hospital Toulouse, 31300 Toulouse, France;
| | - Jean-Louis Pujol
- Département d’Oncologie Thoracique, CHU Montpellier, Univ. Montpellier, 34595 Montpellier, France;
| | - Alexandra Langlais
- Intergroupe Francophone de Cancérologie Thoracique (IFCT), 75009 Paris, France; (A.L.); (F.M.)
| | - Franck Morin
- Intergroupe Francophone de Cancérologie Thoracique (IFCT), 75009 Paris, France; (A.L.); (F.M.)
| | - Gérard Zalcman
- U830 INSERM “Genetics and Biology of Cancers, A.R.T Group”, Curie Institute, 75005 Paris, France
- Department of Thoracic Oncology & CIC1425, Hôpital Bichat-Claude Bernard, Assistance Publique Hôpitaux de Paris, Université Paris-Diderot, 75018 Paris, France
- Correspondence: (G.Z.); (G.L.); Tel.: +33-(0)140-257-502 (G.Z.); +33-(0)231-063-134 (G.L.)
| | - Guénaëlle Levallet
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP CYCERON, 14074 Caen, France; (F.D.); (M.K.); (E.M.); (E.B.)
- Department of Pathology, CHU de Caen, 14033 Caen, France
- Correspondence: (G.Z.); (G.L.); Tel.: +33-(0)140-257-502 (G.Z.); +33-(0)231-063-134 (G.L.)
| |
Collapse
|
8
|
El Gaafary M, Hafner S, Lang SJ, Jin L, Sabry OM, Vogel CV, Vanderwal CD, Syrovets T, Simmet T. A Novel Polyhalogenated Monoterpene Induces Cell Cycle Arrest and Apoptosis in Breast Cancer Cells. Mar Drugs 2019; 17:md17080437. [PMID: 31349625 PMCID: PMC6723102 DOI: 10.3390/md17080437] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 07/17/2019] [Accepted: 07/23/2019] [Indexed: 12/13/2022] Open
Abstract
Breast cancer is the most common cancer type and a primary cause of cancer mortality among females worldwide. Here, we analyzed the anticancer efficacy of a novel bromochlorinated monoterpene, PPM1, a synthetic analogue of polyhalogenated monoterpenes from Plocamium red algae and structurally similar non-brominated monoterpenes. PPM1, but not the non-brominated monoterpenes, decreased selectively the viability of several triple-negative as well as triple-positive breast cancer cells with different p53 status without significantly affecting normal breast epithelial cells. PPM1 induced accumulation of triple-negative MDA-MB-231 cells with 4N DNA content characterized by decreased histone H3-S10/T3 phosphorylation indicating cell cycle arrest in the G2 phase. Western immunoblot analysis revealed that PPM1 treatment triggered an initial rapid activation of Aurora kinases A/B/C and p21Waf1/Cip1 accumulation, which was followed by accumulation of polyploid >4N cells. Flow cytometric analysis showed mitochondrial potential disruption, caspase 3/7 activation, phosphatidylserine externalization, reduction of the amount polyploid cells, and DNA fragmentation consistent with induction of apoptosis. Cell viability was partially restored by the pan-caspase inhibitor Z-VAD-FMK indicating caspase contribution. In vivo, PPM1 inhibited growth, proliferation, and induced apoptosis in MDA-MB-231 xenografted onto the chick chorioallantoic membrane. Hence, Plocamium polyhalogenated monoterpenes and synthetic analogues deserve further exploration as promising anticancer lead compounds.
Collapse
Affiliation(s)
- Menna El Gaafary
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Susanne Hafner
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany
| | - Sophia J Lang
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany
| | - Lu Jin
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany
| | - Omar M Sabry
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Carl V Vogel
- Department of Chemistry, 1102 Natural Sciences II, University of California, Irvine, CA 92697-2025, USA
| | - Christopher D Vanderwal
- Department of Chemistry, 1102 Natural Sciences II, University of California, Irvine, CA 92697-2025, USA
| | - Tatiana Syrovets
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany.
| | - Thomas Simmet
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, D-89081 Ulm, Germany.
| |
Collapse
|
9
|
Fekete JT, Győrffy B. ROCplot.org: Validating predictive biomarkers of chemotherapy/hormonal therapy/anti-HER2 therapy using transcriptomic data of 3,104 breast cancer patients. Int J Cancer 2019; 145:3140-3151. [PMID: 31020993 DOI: 10.1002/ijc.32369] [Citation(s) in RCA: 227] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 04/02/2019] [Accepted: 04/17/2019] [Indexed: 11/10/2022]
Abstract
Systemic therapy of breast cancer can include chemotherapy, hormonal therapy and targeted therapy. Prognostic biomarkers are able to predict survival and predictive biomarkers are able to predict therapy response. In this report, we describe the initial release of the first available online tool able to identify gene expression-based predictive biomarkers using transcriptomic data of a large set of breast cancer patients. Published gene expression data of 36 publicly available datasets were integrated with treatment data into a unified database. Response to therapy was determined using either author-reported pathological complete response data (n = 1,775) or relapse-free survival status at 5 years (n = 1,329). Treatment data includes chemotherapy (n = 2,108), endocrine therapy (n = 971) and anti-human epidermal growth factor receptor 2 (HER2) therapy (n = 267). The transcriptomic database includes 20,089 unique genes and 54,675 probe sets. Gene expression and therapy response are compared using receiver operating characteristics and Mann-Whitney tests. We demonstrate the utility of the pipeline by cross-validating 23 paclitaxel resistance-associated genes in different molecular subtypes of breast cancer. An additional set of established biomarkers including TP53 for chemotherapy in Luminal breast cancer (p = 1.01E-19, AUC = 0.769), HER2 for trastuzumab therapy (p = 8.4E-04, AUC = 0.629) and PGR for hormonal therapy (p = 8.6E-05, AUC = 0.7), are also endorsed. The tool is designed to validate and rank new predictive biomarker candidates in real time. By analyzing the selected genes in a large set of independent patients, one can select the most robust candidates and quickly eliminate those that are most likely to fail in a clinical setting. The analysis tool is accessible at www.rocplot.org.
Collapse
Affiliation(s)
- János T Fekete
- 2nd Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Balázs Győrffy
- 2nd Department of Pediatrics, Semmelweis University, Budapest, Hungary.,MTA TTK Lendület Cancer Biomarker Research Group, Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary
| |
Collapse
|
10
|
Franco MS, Roque MC, Oliveira MC. Short and Long-Term Effects of the Exposure of Breast Cancer Cell Lines to Different Ratios of Free or Co-Encapsulated Liposomal Paclitaxel and Doxorubicin. Pharmaceutics 2019; 11:pharmaceutics11040178. [PMID: 30979090 PMCID: PMC6523953 DOI: 10.3390/pharmaceutics11040178] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 03/29/2019] [Accepted: 03/31/2019] [Indexed: 12/15/2022] Open
Abstract
Background: Associating paclitaxel (PTX) to doxorubicin (DXR) is one of the main chemotherapy strategies for breast cancer (BC) management. Protocols currently available consist in administering both drugs on their maximum tolerated dose, not taking into account the possible differences in efficacy due to their combination ratio. In the present study, the short and long-term cytotoxic effects as well as migratory effects of PTX, DXR, and its combinations at 10:1; 1:1 and 1:10 PTX:DXR molar ratios either free or co-encapsulated in liposomes were evaluated against three human BC cell lines (MDA-MB-231, MCF-7, and SKBR-3). Method: The MTT assay was used to screen for synergy or antagonism between PTX and DXR and the combination index value was calculated using the CalcuSyn software. Nuclear morphological alterations were evaluated by staining the cells with Hoescht 33342. The investigation of senescence and clonogenicity of BC cell lines exposed to different treatments was also studied. In addition, the ability of these cells to migrate was assessed. Results: Taken together, the results presented herein allow us to suggest that there is no benefit in enhancing the PTX concentration above that of DXR in the combination for any of the three cell lines tested. Conclusion: The developed liposomes co-encapsulating PTX and DXR in different molar ratios retained the biological properties of the mixture of free drugs and are valuable for planning new therapeutic strategies.
Collapse
Affiliation(s)
- Marina Santiago Franco
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil.
| | - Marjorie Coimbra Roque
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil.
| | - Mônica Cristina Oliveira
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
11
|
Nuclear Imaging Study of the Pharmacodynamic Effects of Debio 1143, an Antagonist of Multiple Inhibitor of Apoptosis Proteins (IAPs), in a Triple-Negative Breast Cancer Model. CONTRAST MEDIA & MOLECULAR IMAGING 2019; 2018:8494031. [PMID: 30627061 PMCID: PMC6305031 DOI: 10.1155/2018/8494031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 10/18/2018] [Indexed: 11/17/2022]
Abstract
Background Debio 1143, a potent orally available SMAC mimetic, targets inhibitors of apoptosis proteins (IAPs) members and is currently in clinical trials. In this study, nuclear imaging evaluated the effects of Debio 1143 on tumor cell death and metabolism in a triple-negative breast cancer (TNBC) cell line (MDA-MB-231)-based animal model. Methods Apoptosis induced by Debio 1143 was assessed by FACS (caspase-3, annexin 5 (A5)), binding of 99mTc-HYNIC-Annexin V, and a cell proliferation assay. 99mTc-HYNIC-Annexin V SPECT and [18F]-FDG PET were also performed in mice xenografted with MDA-MB-231 cells. Results Debio 1143 induced early apoptosis both in vitro and in vivo 6 h after treatment. Debio 1143 inhibited tumor growth, which was associated with a decreased tumor [18F]-FDG uptake when measured during treatment. Conclusions This imaging study combining SPECT and PET showed the early proapoptotic effects of Debio 1143 resulting in a robust antitumor activity in a preclinical TNBC model. These imaging biomarkers represent valuable noninvasive tools for translational and clinical research in TNBC.
Collapse
|
12
|
Dzobo K, Hassen N, Senthebane DA, Thomford NE, Rowe A, Shipanga H, Wonkam A, Parker MI, Mowla S, Dandara C. Chemoresistance to Cancer Treatment: Benzo-α-Pyrene as Friend or Foe? Molecules 2018; 23:E930. [PMID: 29673198 PMCID: PMC6017867 DOI: 10.3390/molecules23040930] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 04/13/2018] [Accepted: 04/15/2018] [Indexed: 12/14/2022] Open
Abstract
Background: Environmental pollution such as exposure to pro-carcinogens including benzo-α-pyrene is becoming a major problem globally. Moreover, the effects of benzo-α-pyrene (BaP) on drug pharmacokinetics, pharmacodynamics, and drug resistance warrant further investigation, especially in cancer outpatient chemotherapy where exposure to environmental pollutants might occur. Method: We report here on the effects of benzo-α-pyrene on esophageal cancer cells in vitro, alone, or in combination with chemotherapeutic drugs cisplatin, 5-flurouracil, or paclitaxel. As the study endpoints, we employed expression of proteins involved in cell proliferation, drug metabolism, apoptosis, cell cycle analysis, colony formation, migration, and signaling cascades in the WHCO1 esophageal cancer cell line after 24 h of treatment. Results: Benzo-α-pyrene had no significant effect on WHCO1 cancer cell proliferation but reversed the effect of chemotherapeutic drugs by reducing drug-induced cell death and apoptosis by 30−40% compared to drug-treated cells. The three drugs significantly reduced WHCO1 cell migration by 40−50% compared to control and BaP-treated cells. Combined exposure to drugs was associated with significantly increased apoptosis and reduced colony formation. Evaluation of survival signaling cascades showed that although the MEK-ERK and Akt pathways were activated in the presence of drugs, BaP was a stronger activator of the MEK-ERK and Akt pathways than the drugs. Conclusion: The present study suggest that BaP can reverse the effects of drugs on cancer cells via the activation of survival signaling pathways and upregulation of anti-apoptotic proteins such as Bcl-2 and Bcl-xL. Our data show that BaP contribute to the development of chemoresistant cancer cells.
Collapse
Affiliation(s)
- Kevin Dzobo
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Wernher and Beit Building (South), University of Cape Town Medical Campus, Anzio Road, Observatory 7925, Cape Town, South Africa.
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa.
| | - Naseeha Hassen
- Pharmacogenomics and Drug Metabolism Group, Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa.
| | - Dimakatso Alice Senthebane
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Wernher and Beit Building (South), University of Cape Town Medical Campus, Anzio Road, Observatory 7925, Cape Town, South Africa.
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa.
| | - Nicholas Ekow Thomford
- Pharmacogenomics and Drug Metabolism Group, Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa.
| | - Arielle Rowe
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Wernher and Beit Building (South), University of Cape Town Medical Campus, Anzio Road, Observatory 7925, Cape Town, South Africa.
| | - Hendrina Shipanga
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Wernher and Beit Building (South), University of Cape Town Medical Campus, Anzio Road, Observatory 7925, Cape Town, South Africa.
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa.
| | - Ambroise Wonkam
- Pharmacogenomics and Drug Metabolism Group, Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa.
| | - M Iqbal Parker
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa.
| | - Shaheen Mowla
- Division of Haematology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa.
| | - Collet Dandara
- Pharmacogenomics and Drug Metabolism Group, Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa.
| |
Collapse
|
13
|
Duan Z, Chen C, Qin J, Liu Q, Wang Q, Xu X, Wang J. Cell-penetrating peptide conjugates to enhance the antitumor effect of paclitaxel on drug-resistant lung cancer. Drug Deliv 2017; 24:752-764. [PMID: 28468542 PMCID: PMC8253140 DOI: 10.1080/10717544.2017.1321060] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/13/2017] [Accepted: 04/17/2017] [Indexed: 01/01/2023] Open
Abstract
To conquer the drug resistance of tumors and the poor solubility of paclitaxel (PTX), two PTX-cell-penetrating peptide conjugates (PTX-CPPs), PTX-TAT and PTX-LMWP, were synthesized and evaluated for the first time. Compared with free PTX, PTX-CPPs displayed significantly enhanced cellular uptake, elevated cell toxicity, increased cell apoptosis, and decreased mitochondrial membrane potential (Δψm) in both A549 and A549T cells. PTX-LMWP exhibited a stronger inhibitory effect than PTX-TAT in A549T cells. Analysis of cell-cycle distribution showed that PTX-LMWP influenced mitosis in drug-resistant A549T tumor cells via a different mechanism than PTX. PTX-CPPs were more efficient in inhibiting tumor growth in tumor-bearing mice than free PTX, which suggested their better in vivo antitumor efficacy. Hence, this study demonstrates that PTX-CPPs, particularly PTX-LMWP, have outstanding potential for inhibiting the growth of tumors and are a promising approach for treating lung cancer, especially drug-resistant lung cancer.
Collapse
Affiliation(s)
- Ziqing Duan
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, PR China
| | - Cuitian Chen
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, PR China
| | - Jing Qin
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, PR China
| | - Qi Liu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, PR China
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Traditional Chinese Medicine, Guangzhou, PR China, and
| | - Xinchun Xu
- Shanghai Xuhui Central Hospital, Shanghai, PR China
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, PR China
| |
Collapse
|
14
|
Wang S, Ding M, Chen X, Chang L, Sun Y. Development of bimolecular fluorescence complementation using rsEGFP2 for detection and super-resolution imaging of protein-protein interactions in live cells. BIOMEDICAL OPTICS EXPRESS 2017; 8:3119-3131. [PMID: 28663931 PMCID: PMC5480454 DOI: 10.1364/boe.8.003119] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/12/2017] [Accepted: 05/12/2017] [Indexed: 05/28/2023]
Abstract
Direct visualization of protein-protein interactions (PPIs) at high spatial and temporal resolution in live cells is crucial for understanding the intricate and dynamic behaviors of signaling protein complexes. Recently, bimolecular fluorescence complementation (BiFC) assays have been combined with super-resolution imaging techniques including PALM and SOFI to visualize PPIs at the nanometer spatial resolution. RESOLFT nanoscopy has been proven as a powerful live-cell super-resolution imaging technique. With regard to the detection and visualization of PPIs in live cells with high temporal and spatial resolution, here we developed a BiFC assay using split rsEGFP2, a highly photostable and reversibly photoswitchable fluorescent protein previously developed for RESOLFT nanoscopy. Combined with parallelized RESOLFT microscopy, we demonstrated the high spatiotemporal resolving capability of a rsEGFP2-based BiFC assay by detecting and visualizing specifically the heterodimerization interactions between Bcl-xL and Bak as well as the dynamics of the complex on mitochondria membrane in live cells.
Collapse
Affiliation(s)
- Sheng Wang
- State Key Laboratory of Membrane Biology, Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
- Contributed equally to this work
| | - Miao Ding
- State Key Laboratory of Membrane Biology, Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
- Contributed equally to this work
| | - Xuanze Chen
- State Key Laboratory of Membrane Biology, Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
- Contributed equally to this work
| | - Lei Chang
- State Key Laboratory of Membrane Biology, Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
| | - Yujie Sun
- State Key Laboratory of Membrane Biology, Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
15
|
Bioelectrical impedimetric sensor for single cell analysis based on nanoroughened quartz substrate; suitable for cancer therapeutic purposes. J Pharm Biomed Anal 2017; 142:315-323. [PMID: 28531834 DOI: 10.1016/j.jpba.2017.05.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 05/10/2017] [Accepted: 05/12/2017] [Indexed: 12/17/2022]
Abstract
Single cells analysis has been interested in recent decade. Apart from scientific benefits to achieve new biological phenomena in cell study, many diagnostic and therapeutic protocols in non-communicable diseases were introduced by single cell analysis. Moreover, non-invasive methods to maintain the investigated cell for time dependent monitoring has been widely studied because of its importance in some crucial cases such as drug resistance in cancer. Bioelectrical monitoring is one of such methods Although the procedures reported based on electrical probing might not induce cell disruption, indirect connection between recording electrodes and cell membrane (mostly in microfluidic approaches) reduced the quality of response and limited the precision of the results. Here, a bioelectronic sensor for monitoring the effect of anticancer drugs on single breast cancer cells was fabricated based on nano-roughened gold electrodes on a quartz substrate applied direct contacts to cell membrane. Whole of the surface except a microcircle surrounded the sensing region was passivated by overbaked photoresist layer. Cells were dropped on the sensor without the assistance of any micropipette or microfluidic systems and just individual regions for attachment of one cell has been opened on the sensing region arrays. MCF-7 cancer cells were time tracked under the effect of Paclitaxel and Mebendazole anti-tubulin drugs in low and high doses. Inducing non regulated depolymerization and polymerization in tubulin structures of the single cancer cells were monitored by the electrical signals recorded before and after drug treatment. Electrical responses of single cells to their incubation with drugs completely reflected their vitality and biological states which were confirmed by confocal imaging. This is one of the first investigation on bioelectrical monitoring of single cell's resistance to anticancer drugs.
Collapse
|
16
|
Anticancer, antimicrobial, spectral, voltammetric and DFT studies with Cu(II) complexes of 2-hydroxy-5-methoxyacetophenone thiosemicarbazone and its N(4)- substituted derivatives. J Organomet Chem 2017. [DOI: 10.1016/j.jorganchem.2016.12.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
17
|
Sayeed MA, Bracci M, Lazzarini R, Tomasetti M, Amati M, Lucarini G, Di Primio R, Santarelli L. Use of potential dietary phytochemicals to target miRNA: Promising option for breast cancer prevention and treatment? J Funct Foods 2017. [DOI: 10.1016/j.jff.2016.11.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
18
|
Lang PY, Nanjangud GJ, Sokolsky-Papkov M, Shaw C, Hwang D, Parker JS, Kabanov AV, Gershon TR. ATR maintains chromosomal integrity during postnatal cerebellar neurogenesis and is required for medulloblastoma formation. Development 2016; 143:4038-4052. [PMID: 27803059 PMCID: PMC5117143 DOI: 10.1242/dev.139022] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 09/16/2016] [Indexed: 02/01/2023]
Abstract
Microcephaly and medulloblastoma may both result from mutations that compromise genomic stability. We report that ATR, which is mutated in the microcephalic disorder Seckel syndrome, sustains cerebellar growth by maintaining chromosomal integrity during postnatal neurogenesis. Atr deletion in cerebellar granule neuron progenitors (CGNPs) induced proliferation-associated DNA damage, p53 activation, apoptosis and cerebellar hypoplasia in mice. Co-deletions of either p53 or Bax and Bak prevented apoptosis in Atr-deleted CGNPs, but failed to fully rescue cerebellar growth. ATR-deficient CGNPs had impaired cell cycle checkpoint function and continued to proliferate, accumulating chromosomal abnormalities. RNA-Seq demonstrated that the transcriptional response to ATR-deficient proliferation was highly p53 dependent and markedly attenuated by p53 co-deletion. Acute ATR inhibition in vivo by nanoparticle-formulated VE-822 reproduced the developmental disruptions seen with Atr deletion. Genetic deletion of Atr blocked tumorigenesis in medulloblastoma-prone SmoM2 mice. Our data show that p53-driven apoptosis and cell cycle arrest - and, in the absence of p53, non-apoptotic cell death - redundantly limit growth in ATR-deficient progenitors. These mechanisms may be exploited for treatment of CGNP-derived medulloblastoma using ATR inhibition.
Collapse
Affiliation(s)
- Patrick Y Lang
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Neurology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Gouri J Nanjangud
- Molecular Cytogenetics Core Facility, Memorial Sloan Kettering Research Center, New York, NY 10021, USA
| | - Marina Sokolsky-Papkov
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Christine Shaw
- Molecular Cytogenetics Core Facility, Memorial Sloan Kettering Research Center, New York, NY 10021, USA
| | - Duhyeong Hwang
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Joel S Parker
- Department of Genetics, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Alexander V Kabanov
- Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Timothy R Gershon
- Department of Neurology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
19
|
He Y, Yan D, Zheng D, Hu Z, Li H, Li J. Cell Division Cycle 6 Promotes Mitotic Slippage and Contributes to Drug Resistance in Paclitaxel-Treated Cancer Cells. PLoS One 2016; 11:e0162633. [PMID: 27611665 PMCID: PMC5017606 DOI: 10.1371/journal.pone.0162633] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 08/25/2016] [Indexed: 12/18/2022] Open
Abstract
Paclitaxel (PTX) is an antimitotic drug that possesses potent anticancer activity, but its therapeutic potential in the clinic has been hindered by drug resistance. Here, we report a mechanism by which cancer cells can exit from the PTX-induced mitotic arrest, i.e. mitotic slippage, and avoid subsequent death resulting in drug resistance. In cells experiencing mitotic slippage, Cdc6 protein level was significantly upregulated, Cdk1 activity was inhibited, and Cohesin/Rad21 was cleaved as a result. Cdc6 depletion by RNAi or Norcantharidin inhibited PTX-induced Cdc6 up-regulation, maintained Cdk1 activity, and repressed Cohesin/Rad21 cleavage. In all, this resulted in reduced mitotic slippage and reversal of PTX resistance. Moreover, in synchronized cells, the role of Cdc6 in mitotic exit under PTX pressure was also confirmed. This study indicates that Cdc6 may promote mitotic slippage by inactivation of Cdk1. Targeting of Cdc6 may serve as a promising strategy for enhancing the anticancer activity of PTX.
Collapse
Affiliation(s)
- Yue He
- Institute of Biotherapy, School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Daoyu Yan
- Institute of Biotherapy, School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Dianpeng Zheng
- Institute of Biotherapy, School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhiming Hu
- Institute of Biotherapy, School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongwei Li
- Institute of Biotherapy, School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
- * E-mail: (JL); (HL)
| | - Jinlong Li
- Institute of Biotherapy, School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
- * E-mail: (JL); (HL)
| |
Collapse
|
20
|
Gasca J, Flores ML, Giráldez S, Ruiz-Borrego M, Tortolero M, Romero F, Japón MA, Sáez C. Loss of FBXW7 and accumulation of MCL1 and PLK1 promote paclitaxel resistance in breast cancer. Oncotarget 2016; 7:52751-52765. [PMID: 27409838 PMCID: PMC5288146 DOI: 10.18632/oncotarget.10481] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 06/21/2016] [Indexed: 02/07/2023] Open
Abstract
FBXW7 is a component of SCF (complex of SKP1, CUL1 and F-box-protein)-type ubiquitin ligases that targets several oncoproteins for ubiquitination and degradation by the proteasome. FBXW7 regulates cellular apoptosis by targeting MCL1 for ubiquitination. Recently, we identified PLK1 as a new substrate of FBXW7 modulating the intra-S-phase DNA-damage checkpoint. Taxanes are frequently used in breast cancer treatments, but the acquisition of resistance makes these treatments ineffective. We investigated the role of FBXW7 and their substrates MCL1 and PLK1 in regulating the apoptotic response to paclitaxel treatment in breast cancer cells and their expression in breast cancer tissues. Paclitaxel-sensitive MDA-MB-468 and a paclitaxel-resistant MDA-MB-468R subclone were used to study the role of FBXW7 and substrates in paclitaxel-induced apoptosis. Forced expression of FBXW7 or downregulation of MCL1 or PLK1 restored sensitivity to paclitaxel in MDA-MB-468R cells. By contrary, FBXW7-silenced MDA-MB-468 cells became resistant to paclitaxel. The expression of FBXW7 and substrates were studied in 296 invasive carcinomas by immunohistochemistry and disease-free survival was analyzed in a subset of patients treated with paclitaxel. In breast cancer tissues, loss of FBXW7 correlated with adverse prognosis markers and loss of FBXW7 and MCL1 or PLK1 accumulation were associated with diminished disease-free survival in paclitaxel-treated patients. We conclude that FBXW7 regulates the response to paclitaxel by targeting MCL1 and PLK1 in breast cancer cells and thus targeting these substrates may be a valuable adjunct for paclitaxel treatment. Also, FBXW7, MCL1 and PLK1 may be relevant predictive markers of tumor progression and response to paclitaxel treatment.
Collapse
Affiliation(s)
- Jessica Gasca
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Maria Luz Flores
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Servando Giráldez
- Department of Microbiology, Faculty of Biology, Universidad de Sevilla, Seville, Spain
| | | | - María Tortolero
- Department of Microbiology, Faculty of Biology, Universidad de Sevilla, Seville, Spain
| | - Francisco Romero
- Department of Microbiology, Faculty of Biology, Universidad de Sevilla, Seville, Spain
| | - Miguel A. Japón
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Department of Pathology, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Carmen Sáez
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Department of Pathology, Hospital Universitario Virgen del Rocío, Seville, Spain
| |
Collapse
|
21
|
Chiu YJ, Cai W, Shih YRV, Lian I, Lo YH. A Single-Cell Assay for Time Lapse Studies of Exosome Secretion and Cell Behaviors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:3658-66. [PMID: 27254278 PMCID: PMC5023418 DOI: 10.1002/smll.201600725] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 04/18/2016] [Indexed: 05/17/2023]
Abstract
To understand the inhomogeneity of cells in biological systems, there is a growing demand on the capability of characterizing the properties of individual single cells. Since single-cell studies require continuous monitoring of the cell behaviors, an effective single-cell assay that can support time lapsed studies in a high throughput manner is desired. Most currently available single-cell technologies cannot provide proper environments to sustain cell growth and, proliferation of single cells and convenient, noninvasive tests of single-cell behaviors from molecular markers. Here, a highly versatile single-cell assay is presented that can accommodate different cellular types, enable easy and efficient single-cell loading and culturing, and be suitable for the study of effects of in vitro environmental factors in combination with drug screening. One salient feature of the assay is the noninvasive collection and surveying of single-cell secretions at different time points, producing unprecedented insight of single-cell behaviors based on the biomarker signals from individual cells under given perturbations. Above all, the acquired information is quantitative, for example, measured by the number of exosomes each single-cell secretes for a given time period. Therefore, our single-cell assay provides a convenient, low-cost, and enabling tool for quantitative, time lapsed studies of single-cell properties.
Collapse
Affiliation(s)
- Yu-Jui Chiu
- Materials Science and Engineering Program, University of California at San Diego, La Jolla, California, USA
| | - Wei Cai
- Materials Science and Engineering Program, University of California at San Diego, La Jolla, California, USA
| | - Yu-Ru V. Shih
- Department of Bioengineering, University of California at San Diego, La Jolla, California, USA
| | - Ian Lian
- Department of Biology, Lamar University, Beaumont, Texas, USA
| | - Yu-Hwa Lo
- Materials Science and Engineering Program, University of California at San Diego, La Jolla, California, USA
- Department of Electrical and Computer Engineering, University of California at San Diego, La Jolla, California, USA
| |
Collapse
|
22
|
Badr El-Din NK, Ali DA, Alaa El-Dein M, Ghoneum M. Enhancing the Apoptotic Effect of a Low Dose of Paclitaxel on Tumor Cells in Mice by Arabinoxylan Rice Bran (MGN-3/Biobran). Nutr Cancer 2016; 68:1010-20. [PMID: 27367621 DOI: 10.1080/01635581.2016.1192204] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
In this study, we examine the ability of arabinoxylan rice bran (MGN-3/Biobran) to enhance the apoptotic effect of paclitaxel (Taxol) at low concentration [2 mg/kg body weight (BW)] in animals bearing Ehrlich ascites carcinoma (EAC) cells and elucidate its mechanisms of action. On Day 8 following tumor cells inoculation, mice bearing tumors were administered MGN-3 alone (40 mg/kg BW), paclitaxel alone, or MGN-3 plus paclitaxel. On Day 30 post-tumor inoculation, we observed significant suppression of tumor volume (TV) with paclitaxel alone (59%), MGN-3 alone (77%), and MGN-3 plus paclitaxel (88%). Inhibition of tumor growth post-treatment with both agents, as compared with either treatment alone, was associated with a decrease in cell proliferation, a marked increase in the sub-G0/G1 population, an increase in DNA damage and apoptosis of tumor cells, and a significant maximization of the apoptosis index (AI)/proliferation index (PrI) ratio. Histopathological and electron microscopy examination of the combined treatment group showed an increase in the degenerative regions of the solid tumor tissue and abundant apoptotic cells. These data suggest that MGN-3 supplementation enhances tumor cell demise in the presence of a low dose of chemotherapeutic agent via apoptotic mechanism.
Collapse
Affiliation(s)
- Nariman K Badr El-Din
- a Department of Zoology , Faculty of Science, University of Mansoura , Mansoura , Egypt
| | - Doaa A Ali
- a Department of Zoology , Faculty of Science, University of Mansoura , Mansoura , Egypt
| | - Mai Alaa El-Dein
- a Department of Zoology , Faculty of Science, University of Mansoura , Mansoura , Egypt
| | - Mamdooh Ghoneum
- b Department of Otolaryngology , Charles Drew University of Medicine and Science , Los Angeles , California , USA
| |
Collapse
|
23
|
Dai Z, Yao Q, Zhu L. MMP2-Sensitive PEG-Lipid Copolymers: A New Type of Tumor-Targeted P-Glycoprotein Inhibitor. ACS APPLIED MATERIALS & INTERFACES 2016; 8:12661-73. [PMID: 27145021 DOI: 10.1021/acsami.6b03064] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Low tumor targetability and multidrug resistance (MDR) are two major impediments to the success of cancer treatments. Nanomaterials which possess high tumor targetability and the ability to reverse the MDR are rare. This report describes a new type of self-assembling polyethylene glycol-phosphoethanolamine-based copolymers (PEG-pp-PE) which showed both the matrix metalloproteinase 2 (MMP2)-sensitive tumor-targeted drug delivery and ability to inhibit the P-glycoprotein (P-gp)-mediated drug efflux. In this study, we synthesized a series of the homologous analogues of PEG-pp-PE copolymers and investigated the influence of their structures, including PEG lengths and peptide linkers, on the drug efflux, and identified the underlying mechanisms. We found that the whole structure (PEG-peptide-lipid) rather than any parts of the copolymers was key for the P-gp inhibition and a delicate balance between the hydrophilic and lipophilic segments of the PEG-pp-PE copolymers was needed for better modulating the P-gp-mediated drug efflux. The best copolymer, PEG2k-pp-PE, showed even higher P-gp inhibition effect than the d-α-tocopherol polyethylene glycol 1000 succinate (TPGS1k). We also found that the P-gp inhibition capability of PEG-pp-PE copolymers was highly associated with the P-gp down-regulation, the increase in the plasma membrane fluidity, and the inhibition of the P-gp ATPase activity. Besides, the excellent physicochemical properties, high drug loading, MMP2-dependent drug release, and improved drug efficacy in the MDR cancer cells suggested that the PEG-pp-PE copolymers might have great potential for building tumor-targeted drug delivery systems for treating drug-resistant cancers.
Collapse
Affiliation(s)
- Zhi Dai
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Health Science Center , Kingsville, Texas 78363, United States
| | - Qing Yao
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Health Science Center , Kingsville, Texas 78363, United States
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University , Shenyang 110016, Liaoning, People's Republic of China
| | - Lin Zhu
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Health Science Center , Kingsville, Texas 78363, United States
| |
Collapse
|
24
|
Prunier C, Josserand V, Vollaire J, Beerling E, Petropoulos C, Destaing O, Montemagno C, Hurbin A, Prudent R, de Koning L, Kapur R, Cohen PA, Albiges-Rizo C, Coll JL, van Rheenen J, Billaud M, Lafanechère L. LIM Kinase Inhibitor Pyr1 Reduces the Growth and Metastatic Load of Breast Cancers. Cancer Res 2016; 76:3541-52. [DOI: 10.1158/0008-5472.can-15-1864] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 04/03/2016] [Indexed: 11/16/2022]
|
25
|
Yeh CC, Ko HH, Hsieh YP, Wu KJ, Kuo MYP, Deng YT. Phenethyl isothiocyanate enhances TRAIL-induced apoptosis in oral cancer cells and xenografts. Clin Oral Investig 2016; 20:2343-2352. [DOI: 10.1007/s00784-016-1736-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 01/21/2016] [Indexed: 11/29/2022]
|
26
|
Dorman SN, Baranova K, Knoll JHM, Urquhart BL, Mariani G, Carcangiu ML, Rogan PK. Genomic signatures for paclitaxel and gemcitabine resistance in breast cancer derived by machine learning. Mol Oncol 2015; 10:85-100. [PMID: 26372358 DOI: 10.1016/j.molonc.2015.07.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 07/31/2015] [Indexed: 12/21/2022] Open
Abstract
Increasingly, the effectiveness of adjuvant chemotherapy agents for breast cancer has been related to changes in the genomic profile of tumors. We investigated correspondence between growth inhibitory concentrations of paclitaxel and gemcitabine (GI50) and gene copy number, mutation, and expression first in breast cancer cell lines and then in patients. Genes encoding direct targets of these drugs, metabolizing enzymes, transporters, and those previously associated with chemoresistance to paclitaxel (n = 31 genes) or gemcitabine (n = 18) were analyzed. A multi-factorial, principal component analysis (MFA) indicated expression was the strongest indicator of sensitivity for paclitaxel, and copy number and expression were informative for gemcitabine. The factors were combined using support vector machines (SVM). Expression of 15 genes (ABCC10, BCL2, BCL2L1, BIRC5, BMF, FGF2, FN1, MAP4, MAPT, NFKB2, SLCO1B3, TLR6, TMEM243, TWIST1, and CSAG2) predicted cell line sensitivity to paclitaxel with 82% accuracy. Copy number profiles of 3 genes (ABCC10, NT5C, TYMS) together with expression of 7 genes (ABCB1, ABCC10, CMPK1, DCTD, NME1, RRM1, RRM2B), predicted gemcitabine response with 85% accuracy. Expression and copy number studies of two independent sets of patients with known responses were then analyzed with these models. These included tumor blocks from 21 patients that were treated with both paclitaxel and gemcitabine, and 319 patients on paclitaxel and anthracycline therapy. A new paclitaxel SVM was derived from an 11-gene subset since data for 4 of the original genes was unavailable. The accuracy of this SVM was similar in cell lines and tumor blocks (70-71%). The gemcitabine SVM exhibited 62% prediction accuracy for the tumor blocks due to the presence of samples with poor nucleic acid integrity. Nevertheless, the paclitaxel SVM predicted sensitivity in 84% of patients with no or minimal residual disease.
Collapse
Affiliation(s)
- Stephanie N Dorman
- Department of Biochemistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Katherina Baranova
- Department of Biochemistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Joan H M Knoll
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada; Molecular Diagnostics Division, Laboratory Medicine Program, London Health Sciences Centre, ON, Canada; Cytognomix Inc., London, ON, Canada
| | - Brad L Urquhart
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Gabriella Mariani
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maria Luisa Carcangiu
- Department of Diagnostic and Laboratory Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Peter K Rogan
- Department of Biochemistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada; Cytognomix Inc., London, ON, Canada; Department of Computer Science, University of Western Ontario, London, ON, Canada; Department of Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.
| |
Collapse
|
27
|
Coburn JM, Kaplan DL. Engineering Biomaterial-Drug Conjugates for Local and Sustained Chemotherapeutic Delivery. Bioconjug Chem 2015; 26:1212-23. [PMID: 25689115 PMCID: PMC4856894 DOI: 10.1021/acs.bioconjchem.5b00046] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The standard of care for cancer patients includes surgical resection, radiation, and chemotherapy with cytotoxic chemotherapy drugs usually part of the treatment. However, these drugs are commonly associated with cardiotoxicity, ototoxicity, nephrotoxicity, peripheral neuropathy, and myelosuppression. Strategies to deliver cytotoxic chemotherapy drugs while reducing secondary toxicity and increasing tumor dosing would therefore be desirable. This goal can be achieved through the use of controlled release drug carrier systems. The aim of this review is to provide an overview of clinically used drug carrier systems and recently developed approaches for drug-biomaterial conjugation.
Collapse
Affiliation(s)
- Jeannine M. Coburn
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| |
Collapse
|
28
|
Qin CZ, Zhang X, Wu LX, Wen CJ, Hu L, Lv QL, Shen DY, Zhou HH. Advances in molecular signaling mechanisms of β-phenethyl isothiocyanate antitumor effects. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:3311-3322. [PMID: 25798652 DOI: 10.1021/jf504627e] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
β-Phenethyl isothiocyanate (PEITC) is an important phytochemical from cruciferous vegetables and is being evaluated for chemotherapeutic activity in early phase clinical trials. Moreover, studies in cell culture and in animals found that the anticarcinogenic activities of PEITC involved all the major stages of tumor growth: initiation, promotion, and progression. A number of mechanisms have been proposed for the chemopreventive activities of this compound. Here, we focus on the major molecular signaling pathways for the anticancer activities of PEITC. These include (1) activation of apoptosis pathways; (2) induction of cell cycle arrest; and (3) inhibition of the survival pathways. Furthermore, we also discussed the regulation of drug-metabolizing enzymes, including cytochrome P450s, metabolizing enzymes, and multidrug resistance.
Collapse
Affiliation(s)
- Chong-Zhen Qin
- †Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, P. R. China
- ‡Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, P. R. China
| | - Xue Zhang
- §Institute of Life Sciences, Chongqing Medical University, Chongqing, Chongqing 400016, China
| | - Lan-Xiang Wu
- §Institute of Life Sciences, Chongqing Medical University, Chongqing, Chongqing 400016, China
| | - Chun-Jie Wen
- §Institute of Life Sciences, Chongqing Medical University, Chongqing, Chongqing 400016, China
| | - Lei Hu
- †Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, P. R. China
- ‡Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, P. R. China
| | - Qiao-Li Lv
- †Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, P. R. China
- ‡Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, P. R. China
| | - Dong-Ya Shen
- †Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, P. R. China
- ‡Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, P. R. China
| | - Hong-Hao Zhou
- †Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, P. R. China
- ‡Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, P. R. China
| |
Collapse
|
29
|
Zhang YJ, Zhan X, Wang L, Ho RJY, Sasaki T. pH-responsive artemisinin dimer in lipid nanoparticles are effective against human breast cancer in a xenograft model. J Pharm Sci 2015; 104:1815-24. [PMID: 25753991 DOI: 10.1002/jps.24407] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 12/16/2014] [Accepted: 01/28/2015] [Indexed: 01/09/2023]
Abstract
Artemisinin (ART), a well-known antimalaria drug, also exhibits anticancer activities. We previously reported a group of novel dimeric artemisinin piperazine conjugates (ADPs) possessing pH-dependent aqueous solubility and a proof-of-concept lipid nanoparticle formulation based on natural egg phosphatidylcholine (EPC). EPC may induce allergic reactions in individuals sensitive to egg products. Therefore, the goal of this report is to develop ADP-synthetic lipid particles suitable for in vivo evaluation. We found that ADP binds to 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) with greater than 90% efficiency and forms drug-lipid particles (d ∼ 80 nm). Cryo-electron microscopy of the ADP drug-lipid particles revealed unilamellar vesicle-like structures. Detailed characterization studies show insertion of the ADP lead compound, ADP109, into the DPPC membrane and the presence of an aqueous core. Over 50% of the ADP109 was released in 48 hours at pH4 compared with less than 20% at neutral. ADP109-lipid particles exhibited high potency against human breast cancer, but was tolerated well by nontumorigenic cells. In MDA-MB-231 mouse xenograft model, lipid-bound ADP109 particles were more effective than paclitaxel in controlling tumor growth. Cellular uptake studies showed endocytosis of the nanoparticles and release of core-trapped marker throughout the cytosol at 37°C. These results demonstrate, for the first time, the in vivo feasibility of lipid-bound ART dimer for cancer chemotherapy.
Collapse
Affiliation(s)
- Yitong J Zhang
- Department of Chemistry, University of Washington, Seattle, Washington, 98195
| | | | | | | | | |
Collapse
|
30
|
Jelínek M, Balušíková K, Schmiedlová M, Němcová-Fürstová V, Šrámek J, Stančíková J, Zanardi I, Ojima I, Kovář J. The role of individual caspases in cell death induction by taxanes in breast cancer cells. Cancer Cell Int 2015; 15:8. [PMID: 25685064 PMCID: PMC4329194 DOI: 10.1186/s12935-015-0155-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 12/31/2014] [Indexed: 12/04/2022] Open
Abstract
Background In previous study we showed that caspase-2 plays the role of an apical caspase in cell death induction by taxanes in breast cancer cells. This study deals with the role of other caspases. We tested breast cancer cell lines SK-BR-3 (functional caspase-3) and MCF-7 (nonfunctional caspase-3). Methods and results Using western blot analysis we demonstrated the activation of initiator caspase-8 and -9 as well as executioner caspase-6 and -7 in both tested cell lines after application of taxanes (paclitaxel, SB-T-1216) at death-inducing concentrations. Caspase-3 activation was also found in SK-BR-3 cells. Employing specific siRNAs after taxane application, suppression of caspase-3 expression significantly increased the number of surviving SK-BR-3 cells. Inhibition of caspase-7 expression also increased the number of surviving SK-BR-3 and MCF-7 cells. On the other hand, suppression of caspase-8 and caspase-9 expression had no significant effect on cell survival. However, caspase-9 seemed to be involved in the activation of caspase-3 and caspase-7. Caspase-3 and caspase-7 appeared to activate mutually. Furthermore, we observed a significant decrease in mitochondrial membrane potential (flow cytometric analysis) and cytochrome c release (confocal microscopy, western blot after cell fractionation) from mitochondria in SK-BR-3 cells. No such changes were observed in MCF-7 cells after taxane treatment. Conclusion We conclude that the activation of apical caspase-2 results in the activation of caspase-3 and -7 without the involvement of mitochondria. Caspase-9 can be activated directly via caspase-2 or alternatively after cytochrome c release from mitochondria. Subsequently, caspase-9 activation can also lead to caspase-3 and -7 activations. Caspase-3 and caspase-7 activate mutually. It seems that there is also a parallel pathway involving mitochondria that can cooperate in taxane-induced cell death in breast cancer cells.
Collapse
Affiliation(s)
- Michael Jelínek
- Department of Cell and Molecular Biology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Kamila Balušíková
- Department of Cell and Molecular Biology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Martina Schmiedlová
- Department of Cell and Molecular Biology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Vlasta Němcová-Fürstová
- Department of Cell and Molecular Biology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jan Šrámek
- Department of Cell and Molecular Biology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jitka Stančíková
- Department of Cell and Molecular Biology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Ilaria Zanardi
- Institute of Chemical Biology and Drug Discovery, State University of New York at Stony Brook, Stony Brook, NY USA
| | - Iwao Ojima
- Institute of Chemical Biology and Drug Discovery, State University of New York at Stony Brook, Stony Brook, NY USA
| | - Jan Kovář
- Department of Cell and Molecular Biology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
31
|
Serna A, Galán-Cobo A, Rodrigues C, Sánchez-Gomar I, Toledo-Aral JJ, Moura TF, Casini A, Soveral G, Echevarría M. Functional inhibition of aquaporin-3 with a gold-based compound induces blockage of cell proliferation. J Cell Physiol 2014; 229:1787-801. [PMID: 24676973 DOI: 10.1002/jcp.24632] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 03/24/2014] [Indexed: 12/13/2022]
Abstract
AQP3 has been correlated with higher transport of glycerol, increment of ATP content, and larger proliferation capacity. Recently, we described the gold(III) complex Auphen as a very selective and potent inhibitor of AQP3's glycerol permeability (Pgly ). Here we evaluated Auphen effect on the proliferation of various mammalian cell lines differing in AQP3 expression level: no expression (PC12), moderate (NIH/3T3) or high (A431) endogenous expression, cells stably expressing AQP3 (PC12-AQP3), and human HEK293T cells transiently transfected (HEK-AQP3) for AQP3 expression. Proliferation was evaluated in the absence or presence of Auphen (5 μM) by counting number of viable cells and analyzing 5-bromo-2'-deoxyuridine (BrdU) incorporation. Auphen reduced ≈50% the proliferation in A431 and PC12-AQP3, ≈15% in HEK-AQP3 and had no effect in PC12-wt and NIH/3T3. Strong arrest in the S-G2/M phases of the cell cycle, supported by analysis of cyclins (A, B1, D1, E) levels, was observed in AQP3-expressing cells treated with Auphen. Flow-cytometry of propidium iodide incorporation and measurements of mitochondrial dehydrogenases activity confirmed absence of cytotoxic effect of the drug. Functional studies evidenced ≈50% inhibition of A431 Pgly by Auphen, showing that the compound's antiproliferative effect correlates with its ability to inhibit AQP3 Pgly . Role of Cys-40 on AQP3 permeability blockage by Auphen was confirmed by analyzing the mutated protein (AQP3-Ser-40). Accordingly, cells transfected with mutated AQP3 gained resistance to the antiproliferative effect of Auphen. These results highlight an Auphen inhibitory effect on proliferation of cells expressing AQP3 and suggest a targeted therapeutic effect on carcinomas with large AQP3 expression.
Collapse
Affiliation(s)
- Ana Serna
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla (Departamento de Fisiología Médica y Biofísica), Seville, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Ando M, Yoshikawa K, Iwase Y, Ishiura S. Usefulness of monitoring γ-H2AX and cell cycle arrest in HepG2 cells for estimating genotoxicity using a high-content analysis system. ACTA ACUST UNITED AC 2014; 19:1246-54. [PMID: 24980598 DOI: 10.1177/1087057114541147] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Formation of the phosphorylated protein γ-H2AX is a well-established marker of DNA strand breakage induced by DNA-damaging compounds. Many of these genotoxic compounds also inhibit cell division, leading to arrest at specific points in the cell cycle. Detection of γ-H2AX in combination with cell cycle arrest may therefore be useful for estimating the genotoxicity of experimental compounds. In this study, we examined γ-H2AX formation and cell cycle arrest using high-content screening (HCS) as a method for determining genotoxicity. HepG2 cells were treated with a panel of compounds and then stained with Hoechst 33342 and anti-γ-H2AX, anti-phospho-histone H3, and anti-tubulin antibodies. In total, 19 genotoxic and 7 nongenotoxic compounds were tested in this study. γ-H2AX production was observed within 1 h posttreatment for the majority of Ames-positive compounds, topoisomerase inhibitors, and DNA polymerase inhibitors. Cell cycle arrest in either the S or G2 phase was detected for all DNA-damaging compounds 24 h posttreatment, whereas tubulin-targeting compounds were shown to induce cell cycle arrest in the mitotic phase. Together, these results show that HCS is a simple, rapid, and effective tool for estimating the genotoxicity of compounds through detection of γ-H2AX production and cell cycle arrest.
Collapse
Affiliation(s)
- Masamitsu Ando
- Safety Research Laboratories, Mitsubishi Tanabe Pharma Corporation, Chiba, Japan Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Keisuke Yoshikawa
- Safety Research Laboratories, Mitsubishi Tanabe Pharma Corporation, Chiba, Japan
| | - Yumiko Iwase
- Safety Research Laboratories, Mitsubishi Tanabe Pharma Corporation, Chiba, Japan
| | - Shoichi Ishiura
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
33
|
Namgung R, Mi Lee Y, Kim J, Jang Y, Lee BH, Kim IS, Sokkar P, Rhee YM, Hoffman AS, Kim WJ. Poly-cyclodextrin and poly-paclitaxel nano-assembly for anticancer therapy. Nat Commun 2014; 5:3702. [DOI: 10.1038/ncomms4702] [Citation(s) in RCA: 176] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Accepted: 03/21/2014] [Indexed: 01/06/2023] Open
|
34
|
Fang L, Cheng Q, Bai J, Qi YD, Liu JJ, Li LT, Zheng JN. An oncolytic adenovirus expressing interleukin-24 enhances antitumor activities in combination with paclitaxel in breast cancer cells. Mol Med Rep 2013; 8:1416-24. [PMID: 24042845 DOI: 10.3892/mmr.2013.1680] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 08/21/2013] [Indexed: 11/06/2022] Open
Abstract
Oncolytic adenoviruses are a novel class of anticancer treatment, based upon their ability to replicate selectively within malignant cells resulting in cell lysis. The replication‑selective adenovirus, ZD55‑IL‑24, was constructed by harboring an E1B‑55 kDa deletion and arming with interleukin-24 (IL-24). The microtubule‑stabilizing drug paclitaxel (PTX) exhibits activity in relapsed cancer. In the present study, the synergistic antitumor effects of the combination of PTX and ZD55‑IL‑24 on breast cancer cells was investigated. The results demonstrated that there were different roles for PTX in the expression of transgenic mRNA and protein. ZD55‑IL‑24 combined with PTX induced marked growth inhibition of MDA‑MB‑231 and Bcap‑37 cells. PTX increased viral uptake and appeared not to alter the replication of ZD55‑IL‑24 in breast cancer cells. Annexin V‑fluorescein isothiocyanate/propidium iodide staining and the Hoechst 33258 assay indicated that ZD55‑IL‑24 induced an increase in the number of apoptotic cells when administered in combination with PTX. It was demonstrated that ZD55‑IL‑24 conjugated with PTX was highly concomitant, and increased proapoptotic proteins levels, activated caspase‑3, -7 and -9 and downregulated anti‑apoptotic proteins. These results suggested that ZD55‑IL‑24 in combination with PTX exhibited a markedly increased cytotoxic and apoptosis‑inducing effect in breast cancer cells. Thus, this chemo‑gene‑viro therapeutic strategy was demonstrated to be superior to conventional chemotherapy or gene‑viro therapy alone.
Collapse
Affiliation(s)
- Lin Fang
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | | | | | | | | | | | | |
Collapse
|
35
|
Lainé AL, Adriaenssens E, Vessières A, Jaouen G, Corbet C, Desruelles E, Pigeon P, Toillon RA, Passirani C. The in vivo performance of ferrocenyl tamoxifen lipid nanocapsules in xenografted triple negative breast cancer. Biomaterials 2013; 34:6949-56. [DOI: 10.1016/j.biomaterials.2013.05.065] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 05/24/2013] [Indexed: 01/31/2023]
|