1
|
Shim S, Reinacher-Schick A, Kraeft AL, Per Pfeiffer, Tarpgaard LS, Ettrich TJ, Kestler A, Christensen S, Jandu H, Nawabi M, Roest NL, Damstrup L, Vestlev PM, Brünner N, Stenvang J, Ladekarl M. PANTAX: a phase Ib clinical trial of the efflux pump inhibitor SCO-101 in combination with gemcitabine and nab-paclitaxel in non-resectable or metastatic pancreatic cancer. Invest New Drugs 2025; 43:337-347. [PMID: 40272619 PMCID: PMC12048447 DOI: 10.1007/s10637-025-01526-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 03/10/2025] [Indexed: 05/03/2025]
Abstract
De novo or acquired resistance to chemotherapy is ubiquitous in pancreatic ductal adenocarcinoma (PDAC). SCO-101 is an oral compound that may counteract chemo-resistance by interacting with SRPK1, ABCG2 drug transporter, and liver enzyme UGT1A1. We first conducted preclinical experiments in paclitaxel-resistant PDAC cells to access the tumoricidal effects of SCO-101 or SRPK1-inhibitor alone or in combination with paclitaxel. Second, we enrolled 22 patients with non-resectable PDAC in a phase Ib trial to investigate safety and pharmaco-kinetics, and to establish maximum tolerated dose (MTD) by evaluation of dose-limiting toxicities (DLTs) during the first cycle of 80% dose gemcitabine (Gem) and nab-paclitaxel (Nab) together with increasing doses of SCO-101. In paclitaxel-resistant PDAC cells in vitro, a synergistic effect between SCO-101 and paclitaxel was demonstrated. In patients, daily doses for 6 days of SCO-101 resulted in a two- to threefold drug accumulation, and drug exposure was dose proportional. Treatment was well tolerated. Transiently increased blood bilirubin attributable to SCO-101 was observed in 12 cases (55%) and associated with jaundice in three patients. One and two DLTs, respectively, were observed at 150 and 250mg dosing-levels of SCO-101, and the MTD was determined to be 200 mg of SCO-101 daily for 6 days on a bi-weekly schedule together with 80% dose of Gem and Nab. Median progression-free and overall survival was 3.3 and 9.5 months, respectively. In PDAC, SCO-101 can be added to Gem and Nab with little and manageable toxicity. However, no clear added efficacy signal was observed of the combination. Trial registration number: NCT04652205 (Nov 29, 2020).
Collapse
Affiliation(s)
- Susy Shim
- Department of Oncology, Clinical Cancer Research Center, Aalborg University Hospital, Hobrovej 18 - 22, 9000, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Anke Reinacher-Schick
- Dept. of Hematology, Oncology and Palliative Care, St. Josef-Hospital, Bochum, Germany
| | - Anna-Lena Kraeft
- Dept. of Hematology, Oncology and Palliative Care, St. Josef-Hospital, Bochum, Germany
| | - Per Pfeiffer
- Department of Oncology, Odense University Hospital, Odense, Denmark
- University of Southern Denmark, Odense, Denmark
| | - Line Schmidt Tarpgaard
- Department of Oncology, Odense University Hospital, Odense, Denmark
- University of Southern Denmark, Odense, Denmark
| | - Thomas Jens Ettrich
- Department of Internal Medicine I, University Hospital of Ulm - Oberer Eselsberg, Ulm, Germany
| | - Angelika Kestler
- Department of Internal Medicine I, University Hospital of Ulm - Oberer Eselsberg, Ulm, Germany
| | - Signe Christensen
- Department of Oncology, Clinical Cancer Research Center, Aalborg University Hospital, Hobrovej 18 - 22, 9000, Aalborg, Denmark
| | | | | | | | | | | | | | | | - Morten Ladekarl
- Department of Oncology, Clinical Cancer Research Center, Aalborg University Hospital, Hobrovej 18 - 22, 9000, Aalborg, Denmark.
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
2
|
Guo K, Li S, Wu X, Xiong H. Nanomedicine in the Diagnosis and Treatment of Pancreatic Cancer. Pharmaceutics 2025; 17:449. [PMID: 40284444 PMCID: PMC12030228 DOI: 10.3390/pharmaceutics17040449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with increasing incidence and mortality rates, highlighting the urgent need for early diagnosis and treatment. However, early diagnosis of PDAC is extremely challenging due to the atypical early symptoms or the absence of noticeable symptoms. As a result, many patients are diagnosed with local metastasis, and even patients who are eligible for surgical resection have a high postoperative recurrence rate. Consequently, chemotherapy remains the primary treatment for PDAC. However, the unique biological characteristics of PDAC not only promote tumor progression and metastasis but also often lead to chemoresistance, a significant barrier to successful treatment. Recently, nanomaterials have garnered significant attention as promising materials for diagnosing and treating PDAC, showing great potential in cancer therapy, imaging, and drug delivery. Novel targeted nanomedicines, which encapsulate chemotherapy drugs and gene therapy products, offer significant advantages in overcoming resistance. These nanomedicines not only provide innovative solutions to the limitations of conventional chemotherapy but also improve the selectivity for cancer cells to enhance therapeutic outcomes. Current research is focused on the development of advanced nanomedicines, such as liposomes, nanotubes, and polymer-lipid hybrid systems, aimed at making chemotherapy more effective and longer lasting. This review provides a detailed overview of various nanomedicines utilized in the diagnosis and treatment of PDAC and outlines future directions for their development and key breakthroughs.
Collapse
Affiliation(s)
| | | | - Xinyu Wu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (K.G.); (S.L.)
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (K.G.); (S.L.)
| |
Collapse
|
3
|
Dastgerdi NK, Dastgerdi NK, Bayraktutan H, Costabile G, Atyabi F, Dinarvand R, Longobardi G, Alexander C, Conte C. Enhancing siRNA cancer therapy: Multifaceted strategies with lipid and polymer-based carrier systems. Int J Pharm 2024; 663:124545. [PMID: 39098747 DOI: 10.1016/j.ijpharm.2024.124545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/06/2024]
Abstract
Cancers are increasing in prevalence and many challenges remain for their treatment, such as chemoresistance and toxicity. In this context, siRNA-based therapeutics have many potential advantages for cancer therapies as a result of their ability to reduce or prevent expression of specific cancer-related genes. However, the direct delivery of naked siRNA is hindered by issues like enzymatic degradation, insufficient cellular uptake, and poor pharmacokinetics. Hence, the discovery of a safe and efficient delivery vehicle is essential. This review explores various lipid and polymer-based delivery systems for siRNA in cancer treatment. Both polymers and lipids have garnered considerable attention as carriers for siRNA delivery. While all of these systems protect siRNA and enhance transfection efficacy, each exhibits its unique strengths. Lipid-based delivery systems, for instance, demonstrate high entrapment efficacy and utilize cost-effective materials. Conversely, polymeric-based delivery systems offer advantages through chemical modifications. Nonetheless, certain drawbacks still limit their usage. To address these limitations, combining different materials in formulations (lipid, polymer, or targeting agent) could enhance pharmaceutical properties, boost transfection efficacy, and reduce side effects. Furthermore, co-delivery of siRNA with other therapeutic agents presents a promising strategy to overcome cancer resistance. Lipid-based delivery systems have been demonstrated to encapsulate many therapeutic agents and with high efficiency, but most are limited in terms of the functionalities they display. In contrast, polymeric-based delivery systems can be chemically modified by a wide variety of routes to include multiple components, such as release or targeting elements, from the same materials backbone. Accordingly, by incorporating multiple materials such as lipids, polymers, and/or targeting agents in RNA formulations it is possible to improve the pharmaceutical properties and therapeutic efficacy while reducing side effects. This review focuses on strategies to improve siRNA cancer treatments and discusses future prospects in this important field.
Collapse
Affiliation(s)
- Nazgol Karimi Dastgerdi
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK; Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Nazanin Karimi Dastgerdi
- Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hulya Bayraktutan
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK
| | | | - Fatemeh Atyabi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614315, Iran
| | - Rassoul Dinarvand
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614315, Iran.
| | | | - Cameron Alexander
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK
| | - Claudia Conte
- Department of Pharmacy, University of Napoli Federico II, Napoli, Italy.
| |
Collapse
|
4
|
Ouellette V, Bouzriba C, Chavez Alvarez AC, Hamel-Côté G, Fortin S. Modification of the phenyl ring B of phenyl 4-(2-oxoimidazolidin-1-yl)benzenesulfonates by pyridinyl moiety leads to novel antimitotics targeting the colchicine-binding site. Bioorg Med Chem Lett 2024; 105:129745. [PMID: 38614151 DOI: 10.1016/j.bmcl.2024.129745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/05/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
A series of 8 novel pyridinyl 4-(2-oxoimidazolidin-1-yl)benzenesulfonates (PYRIB-SOs) were designed, prepared and evaluated for their mechanism of action. PYRIB-SOs were found to have antiproliferative activity in the nanomolar to submicromolar range on several breast cancer cell lines. Moreover, subsequent biofunctional assays indicated that the most potent PYRIB-SOs 1-3 act as antimitotics binding to the colchicine-binding site (C-BS) of α, β-tubulin and that they arrest the cell cycle progression in the G2/M phase. Microtubule immunofluorescence and tubulin polymerisation assay confirm that they disrupt the cytoskeleton through inhibition of tubulin polymerisation as observed with microtubule-destabilising agents. They also show good overall theoretical physicochemical, pharmacokinetic and druglike properties. Overall, these results show that PYRIB-SOs is a new family of promising antimitotics to be further studied in vivo for biopharmaceutical and pharmacodynamic evaluations.
Collapse
Affiliation(s)
- Vincent Ouellette
- Centre de recherche du CHU de Québec-Université Laval, Axe Oncologie, Hôpital Saint-François d'Assise, 10 rue de l'Espinay, Québec, QC, G1L 3L5, Canada; Faculté de pharmacie, Université Laval, Pavillon Ferdinand-Vandry, 1050 avenue de la Médecine, Québec, QC, G1V 0A6, Canada.
| | - Chahrazed Bouzriba
- Centre de recherche du CHU de Québec-Université Laval, Axe Oncologie, Hôpital Saint-François d'Assise, 10 rue de l'Espinay, Québec, QC, G1L 3L5, Canada; Faculté de pharmacie, Université Laval, Pavillon Ferdinand-Vandry, 1050 avenue de la Médecine, Québec, QC, G1V 0A6, Canada.
| | - Atziri Corin Chavez Alvarez
- Centre de recherche du CHU de Québec-Université Laval, Axe Oncologie, Hôpital Saint-François d'Assise, 10 rue de l'Espinay, Québec, QC, G1L 3L5, Canada; Faculté de pharmacie, Université Laval, Pavillon Ferdinand-Vandry, 1050 avenue de la Médecine, Québec, QC, G1V 0A6, Canada; Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval (IUCPQ), 2725 chemin Ste-Foy, Québec, QC, G1V 4G5, Canada.
| | - Geneviève Hamel-Côté
- Centre de recherche du CHU de Québec-Université Laval, Axe Oncologie, Hôpital Saint-François d'Assise, 10 rue de l'Espinay, Québec, QC, G1L 3L5, Canada.
| | - Sébastien Fortin
- Centre de recherche du CHU de Québec-Université Laval, Axe Oncologie, Hôpital Saint-François d'Assise, 10 rue de l'Espinay, Québec, QC, G1L 3L5, Canada; Faculté de pharmacie, Université Laval, Pavillon Ferdinand-Vandry, 1050 avenue de la Médecine, Québec, QC, G1V 0A6, Canada.
| |
Collapse
|
5
|
Nunes M, Bartosch C, Abreu MH, Richardson A, Almeida R, Ricardo S. Deciphering the Molecular Mechanisms behind Drug Resistance in Ovarian Cancer to Unlock Efficient Treatment Options. Cells 2024; 13:786. [PMID: 38727322 PMCID: PMC11083313 DOI: 10.3390/cells13090786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/01/2024] [Indexed: 05/13/2024] Open
Abstract
Ovarian cancer is a highly lethal form of gynecological cancer. This disease often goes undetected until advanced stages, resulting in high morbidity and mortality rates. Unfortunately, many patients experience relapse and succumb to the disease due to the emergence of drug resistance that significantly limits the effectiveness of currently available oncological treatments. Here, we discuss the molecular mechanisms responsible for resistance to carboplatin, paclitaxel, polyadenosine diphosphate ribose polymerase inhibitors, and bevacizumab in ovarian cancer. We present a detailed analysis of the most extensively investigated resistance mechanisms, including drug inactivation, drug target alterations, enhanced drug efflux pumps, increased DNA damage repair capacity, and reduced drug absorption/accumulation. The in-depth understanding of the molecular mechanisms associated with drug resistance is crucial to unveil new biomarkers capable of predicting and monitoring the kinetics during disease progression and discovering new therapeutic targets.
Collapse
Affiliation(s)
- Mariana Nunes
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal; (M.N.); (R.A.)
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Carla Bartosch
- Porto Comprehensive Cancer Center Raquel Seruca (PCCC), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal; (C.B.); (M.H.A.)
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
- Cancer Biology & Epigenetics Group, Research Center of Portuguese Oncology Institute of Porto (CI-IPO-Porto), Health Research Network (RISE@CI-IPO-Porto), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
| | - Miguel Henriques Abreu
- Porto Comprehensive Cancer Center Raquel Seruca (PCCC), Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal; (C.B.); (M.H.A.)
- Department of Medical Oncology, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal
| | - Alan Richardson
- The School of Pharmacy and Bioengineering, Guy Hilton Research Centre, Keele University, Thornburrow Drive, Stoke-on-Trent ST4 7QB, Staffordshire, UK;
| | - Raquel Almeida
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal; (M.N.); (R.A.)
- Biology Department, Faculty of Sciences, University of Porto (FCUP), 4169-007 Porto, Portugal
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal
| | - Sara Ricardo
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal; (M.N.); (R.A.)
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
| |
Collapse
|
6
|
Baba K, Uemura K, Nakazato R, Ijaz F, Takahashi S, Ikegami K. Δ3-tubulin impairs mitotic spindle morphology and increases nuclear size in pancreatic cancer cells. Med Mol Morphol 2024; 57:59-67. [PMID: 37930423 DOI: 10.1007/s00795-023-00373-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 10/14/2023] [Indexed: 11/07/2023]
Abstract
Cancer cell proliferation is affected by post-translational modifications of tubulin. Especially, overexpression or depletion of enzymes for modifications on the tubulin C-terminal region perturbs dynamic instability of the spindle body. Those modifications include processing of C-terminal amino acids of α-tubulin; detyrosination, and a removal of penultimate glutamic acid (Δ2). We previously found a further removal of the third last glutamic acid, which generates so-called Δ3-tubulin. The effects of Δ3-tubulin on spindle integrities and cell proliferation remain to be elucidated. In this study, we investigated the impacts of forced expression of Δ3-tubulin on the structure of spindle bodies and cell division in a pancreatic cancer cell line, PANC-1. Overexpression of HA-tagged Δ3-tubulin impaired the morphology and orientation of spindle bodies during cell division in PANC-1 cells. In particular, spindle bending was most significantly increased. Expression of EGFP-tagged Δ3-tubulin driven by the endogenous promoter of human TUBA1B also deformed and misoriented spindle bodies. Spindle bending and condensation defects were significantly observed by EGFP-Δ3-tubulin expression. Furthermore, EGFP-Δ3-tubulin expression increased the nuclear size in a dose-dependent manner of EGFP-Δ3-tubulin expression. The expression of EGFP-Δ3-tubulin tended to slow down cell proliferation. Taken together, our results demonstrate that Δ3-tubulin affects the spindle integrity and cell division.
Collapse
Affiliation(s)
- Kenta Baba
- Department of Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
- Department of Anatomy and Developmental Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Kenichiro Uemura
- Department of Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Ryota Nakazato
- Department of Anatomy and Developmental Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Faryal Ijaz
- Department of Anatomy and Developmental Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Shinya Takahashi
- Department of Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Koji Ikegami
- Department of Anatomy and Developmental Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan.
| |
Collapse
|
7
|
Amiri A, Dietz C, Rapp A, Cardoso MC, Stark RW. The cyto-linker and scaffolding protein "plectin" mis-localization leads to softening of cancer cells. NANOSCALE 2023; 15:15008-15026. [PMID: 37668423 DOI: 10.1039/d3nr02226a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Discovering tools to prevent cancer progression requires understanding the fundamental differences between normal and cancer cells. More than a decade ago, atomic force microscopy (AFM) revealed cancer cells' softer body compared to their healthy counterparts. Here, we investigated the mechanism underlying the softening of cancerous cells in comparison with their healthy counterparts based on AFM high resolution stiffness tomography and 3D confocal microscopy. We showed microtubules (MTs) network in invasive ductal carcinoma cell cytoskeleton is basally located and segmented for around 400 nm from the cell periphery. Additionally, the cytoskeleton scaffolding protein plectin exhibits a mis-localization from the cytoplasm to the surface of cells in the carcinoma which justifies the dissociation of the MT network from the cell's cortex. Furthermore, the assessment of MTs' persistence length using a worm-like-chain (WLC) model in high resolution AFM images showed lower persistence length of the single MTs in ductal carcinoma compared to that in the normal state. Overall, these tuned mechanics support the invasive cells to ascertain more flexibility under compressive forces in small deformations. These data provide new insights into the structural origins of cancer aids in progression.
Collapse
Affiliation(s)
- Anahid Amiri
- Physics of Surfaces, Institute of Materials Science, Technical University of Darmstadt, Alarich-Weiss-Str. 2, 64287 Darmstadt, Germany.
| | - Christian Dietz
- Physics of Surfaces, Institute of Materials Science, Technical University of Darmstadt, Alarich-Weiss-Str. 2, 64287 Darmstadt, Germany.
| | - Alexander Rapp
- Cell Biology and Epigenetics, Department of Biology, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - M Cristina Cardoso
- Cell Biology and Epigenetics, Department of Biology, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - Robert W Stark
- Physics of Surfaces, Institute of Materials Science, Technical University of Darmstadt, Alarich-Weiss-Str. 2, 64287 Darmstadt, Germany.
| |
Collapse
|
8
|
Hassan MS, Awasthi N, Ponna S, von Holzen U. Nab-Paclitaxel in the Treatment of Gastrointestinal Cancers-Improvements in Clinical Efficacy and Safety. Biomedicines 2023; 11:2000. [PMID: 37509639 PMCID: PMC10377238 DOI: 10.3390/biomedicines11072000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/03/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Taxanes (paclitaxel and docetaxel) are one of the most useful classes of anticancer drugs. Taxanes are highly hydrophobic; therefore, these drugs must be dissolved in organic solvents (polysorbate or Cremophor EL), which contribute to their toxicities. To reduce this toxicity and to enhance their efficacy, novel formulations have been developed. Nanoparticle albumin-bound paclitaxel (nab-paclitaxel) is an albumin-stabilized, Cremophor-free, and water-soluble nanoparticle formulation of paclitaxel. Nab-paclitaxel has better solubility and less infusion-associated toxicity compared to solvent-based paclitaxel. Additionally, nab-paclitaxel can be given at higher doses and concentrations compared with solvent-based paclitaxel. Based on its superior clinical efficacy and safety profile, nab-paclitaxel received FDA approval for metastatic breast cancer (2008) and NSCLC (2011). Among gastrointestinal cancers, it is now approved in the USA for treating patients with metastatic adenocarcinoma of the pancreas as first-line therapy in combination with gemcitabine. Furthermore, several clinical trials have suggested the potential efficacy of nab-paclitaxel as a single agent or in combination with other agents for the treatment of metastatic esophageal, gastric, bowel, and biliary tract cancers. Nab-paclitaxel has been demonstrated to have greater overall response rates (ORR) with enhanced progression-free survival (PFS), overall survival (OS) and a superior safety profile with fewer adverse effects in patients with gastrointestinal tract cancers. This review summarizes the advantages associated with nab-paclitaxel-based regimens in terms of improving clinical efficacy and the safety profile in upper gastrointestinal cancer.
Collapse
Affiliation(s)
- Md Sazzad Hassan
- Department of Surgery, Indiana University School of Medicine, South Bend, IN 46617, USA
- Harper Cancer Research Institute, South Bend, IN 46617, USA
| | - Niranjan Awasthi
- Department of Surgery, Indiana University School of Medicine, South Bend, IN 46617, USA
- Harper Cancer Research Institute, South Bend, IN 46617, USA
| | - Saisantosh Ponna
- Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, IN 46556, USA
| | - Urs von Holzen
- Department of Surgery, Indiana University School of Medicine, South Bend, IN 46617, USA
- Harper Cancer Research Institute, South Bend, IN 46617, USA
- Goshen Center for Cancer Care, Goshen, IN 46526, USA
- Department of Surgery, University of Basel School of Medicine, 4001 Basel, Switzerland
| |
Collapse
|
9
|
Shi Q, Liu C, Huo L, Tao Y, Zhang H. Silencing TUBB3 Expression Destroys the Tegument and Flame Cells of Echinococcus multilocularis Protoscoleces. Animals (Basel) 2022; 12:ani12182471. [PMID: 36139331 PMCID: PMC9495074 DOI: 10.3390/ani12182471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/02/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022] Open
Abstract
Alveolar echinococcosis (AE), caused by infection with the larvae of Echinococcus multilocularis, is a neglected tropical disease and zoonosis that causes remarkable morbidity in humans and has economic importance in the livestock industry worldwide. The growth of this parasite resembles the invasion and proliferation of malignant tumours. Microtubules, especially the β-tubulin subunit in the exposed end, are the targets of many antitumour drugs. However, the role of TUBB3, which is the most studied isotype in solid tumours and is also a marker of biological aggressiveness associated with the modulation of tumour metastatic abilities in the growth and development of platyhelminths, is unknown. In this study, protoscoleces (PSCs) are cultivated in monophasic medium in vitro. Using electroporated short interfering RNA (siRNA), EmTUBB3 knockdown was performed with two EmTUBB3-specific siRNAs (siRNA-1 and siRNA-2). qRT–PCR was performed to detect the expression of TUBB3. PSCs viability and the evagination rate and number of body contractions were quantified under a light microscope. Scanning electron microscopy (SEM) and transmission electron microscopy (TEM) were used to observe the ultra-morphological changes of the parasites. After siRNA interference, the EmTUBB3 expression in E. multilocularis PSCs was significantly reduced. Reduced viability, a decreased evagination rate and a decreased number of body contractions were also documented. In particular, shrinkage and roughness of the tegument were observed. Ultrastructural changes included marked damage to flame cells, cracked cilia structures enclosed in the cell body and ruptured microtubule structures. EmTUBB3 possibly plays a crucial role in tegument and flame cell integrity in E. multilocularis PSCs. Novel drugs targeting this specific beta-tubulin isotype in E. multilocularis are potential methods for disease control and deserve further attention.
Collapse
|
10
|
Rahnama N, Jahangir M, Alesaeid S, Kahrizi MS, Adili A, Mohammed RN, Aslaminabad R, Akbari M, Özgönül AM. Association between microRNAs and chemoresistance in pancreatic cancer: Current knowledge, new insights, and forthcoming perspectives. Pathol Res Pract 2022; 236:153982. [PMID: 35779293 DOI: 10.1016/j.prp.2022.153982] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/27/2022] [Accepted: 06/11/2022] [Indexed: 11/25/2022]
Abstract
Pancreatic duct adenocarcinoma, commonly known as pancreatic cancer (PC), is a cancer-related cause of death due to delayed diagnosis, metastasis, and drug resistance. Patients with PC suffer from incorrect responses to chemotherapy due to inherent and acquired chemical resistance. Numerous studies have shown the mechanism of the effect of chemoresistance on PC, such as genetic and epigenetic changes or the elucidation of signaling pathways. In this regard, microRNAs (miRNAs) have been identified as essential modulators of gene expression in various cellular functions, including chemoresistance. Thus, identifying the underlying link between microRNAs and PC chemoresistance helps determine the exact pathogenesis of PC. This study aims to classify miRNAs and signaling pathways related to PC chemoresistance, suggesting new therapeutic approaches to overcome PC chemoresistance.
Collapse
Affiliation(s)
- Negin Rahnama
- Department of Internal Medicine and Health Services, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Samira Alesaeid
- Department of Internal Medicine and Rheumatology, Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ali Adili
- Senior Adult Oncology Department, Moffitt Cancer Center, University of South Florida, FL, USA; Department of Oncology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rebar N Mohammed
- Medical Laboratory Analysis Department, College of Health Sciences, Cihan University of Sulaimaniya, Kurdistan Region, Iraq; College of Veterinary Medicine, University of Sulaimani, Sulaimaniyah, Iraq
| | - Ramin Aslaminabad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Akbari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ali Mert Özgönül
- Department of Biochemistry, Faculty of Medicine, Ege University, Bornova, Izmir, Turkey.
| |
Collapse
|
11
|
Duly AMP, Kao FCL, Teo WS, Kavallaris M. βIII-Tubulin Gene Regulation in Health and Disease. Front Cell Dev Biol 2022; 10:851542. [PMID: 35573698 PMCID: PMC9096907 DOI: 10.3389/fcell.2022.851542] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 04/07/2022] [Indexed: 11/24/2022] Open
Abstract
Microtubule proteins form a dynamic component of the cytoskeleton, and play key roles in cellular processes, such as vesicular transport, cell motility and mitosis. Expression of microtubule proteins are often dysregulated in cancer. In particular, the microtubule protein βIII-tubulin, encoded by the TUBB3 gene, is aberrantly expressed in a range of epithelial tumours and is associated with drug resistance and aggressive disease. In normal cells, TUBB3 expression is tightly restricted, and is found almost exclusively in neuronal and testicular tissues. Understanding the mechanisms that control TUBB3 expression, both in cancer, mature and developing tissues will help to unravel the basic biology of the protein, its role in cancer, and may ultimately lead to the development of new therapeutic approaches to target this protein. This review is devoted to the transcriptional and posttranscriptional regulation of TUBB3 in normal and cancerous tissue.
Collapse
Affiliation(s)
- Alastair M. P. Duly
- Children’s Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, Randwick, NSW, Australia
| | - Felicity C. L. Kao
- Children’s Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, Randwick, NSW, Australia
- Australian Center for NanoMedicine, UNSW Sydney, Sydney, NSW, Australia
- School of Women and Children’s Health, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Wee Siang Teo
- Children’s Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, Randwick, NSW, Australia
- Australian Center for NanoMedicine, UNSW Sydney, Sydney, NSW, Australia
| | - Maria Kavallaris
- Children’s Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, Randwick, NSW, Australia
- Australian Center for NanoMedicine, UNSW Sydney, Sydney, NSW, Australia
- School of Women and Children’s Health, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
- UNSW RNA Institute, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
12
|
Kato A, Naitoh I, Naiki-Ito A, Hayashi K, Okumura F, Fujita Y, Sano H, Nishi Y, Miyabe K, Inoue T, Hirano A, Takada H, Yoshida M, Hori Y, Natsume M, Kato H, Takahashi S, Kataoka H. Class III β-Tubulin Expression Is of Value in Selecting nab -Paclitaxel and Gemcitabine as First-Line Therapy in Unresectable Pancreatic Cancer. Pancreas 2022; 51:372-379. [PMID: 35695793 DOI: 10.1097/mpa.0000000000002032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES Nab -paclitaxel and gemcitabine (GnP) or FOLFIRINOX (a combination of leucovorin, fluorouracil, irinotecan, and oxaliplatin [FFX]) is currently recognized as the standard first-line regimen for unresectable pancreatic ductal adenocarcinoma (PDAC). Class III β-tubulin (TUBB3) has the potential to predict resistance to taxane in various tumors; therefore, this study aimed to clarify whether TUBB3 is a predictive marker for GnP response. METHODS We retrospectively reviewed 113 patients with PDAC who received GnP or FFX as first-line chemotherapy and examined immunohistochemically the TUBB3 expression in specimens obtained by endoscopic ultrasound-guided fine-needle aspiration. RESULTS High TUBB3 expression was associated with a significantly lower disease control rate ( P = 0.017) and shorter progression-free survival (PFS) ( P = 0.019), and multivariate analysis revealed that TUBB3 expression was an independent variable for PFS in the GnP first-line group ( P = 0.045). In addition, in the FFX first-line group, TUBB3 expression was not correlated with PFS or overall survival (OS). In all 113 patients, TUBB3 expression was not also associated with OS. CONCLUSIONS Class III β-tubulin might be a predictive factor for the response of GnP, but not a prognostic factor for OS, helping the selection of an optimized first-line chemotherapy regimen for unresectable PDAC.
Collapse
Affiliation(s)
- Akihisa Kato
- From the Departments of Gastroenterology and Metabolism
| | - Itaru Naitoh
- From the Departments of Gastroenterology and Metabolism
| | - Aya Naiki-Ito
- Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya
| | | | - Fumihiro Okumura
- Department of Gastroenterology, Gifu Prefectural Tajimi Hospital, Tajimi
| | - Yasuaki Fujita
- Department of Gastroenterology, Gifu Prefectural Tajimi Hospital, Tajimi
| | - Hitoshi Sano
- Department of Gastroenterology, Toyokawa City Hospital, Toyokawa
| | - Yuji Nishi
- Department of Gastroenterology, Toyokawa City Hospital, Toyokawa
| | - Katsuyuki Miyabe
- Department of Gastroenterology, Nagoya Daini Red Cross Hospital, Nagoya
| | - Tadahisa Inoue
- Department of Gastroenterology, Aichi Medical University School of Medicine, Nagakute
| | - Atsuyuki Hirano
- Department of Gastroenterology, Nagoya City West Medical Center, Nagoya
| | - Hiroki Takada
- Department of Gastroenterology, Kasugai Municipal Hospital, Kasugai, Japan
| | | | - Yasuki Hori
- From the Departments of Gastroenterology and Metabolism
| | | | - Hiroyuki Kato
- Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya
| | - Satoru Takahashi
- Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya
| | | |
Collapse
|
13
|
βIII-Tubulin Structural Domains Regulate Mitochondrial Network Architecture in an Isotype-Specific Manner. Cells 2022; 11:cells11050776. [PMID: 35269398 PMCID: PMC8909761 DOI: 10.3390/cells11050776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/12/2022] [Accepted: 02/20/2022] [Indexed: 02/06/2023] Open
Abstract
βIII-tubulin is a neuronal microtubule protein that is aberrantly expressed in epithelial cancers. The microtubule network is implicated in regulating the architecture and dynamics of the mitochondrial network, although the isotype-specific role for β-tubulin proteins that constitute this microtubule network remains unclear. High-resolution electron microscopy revealed that manipulation of βIII-tubulin expression levels impacts the volume and shape of mitochondria. Analysis of the structural domains of the protein identifies that the C-terminal tail of βIII-tubulin, which distinguishes this protein from other β-tubulin isotypes, significantly contributes to the isotype-specific effects of βIII-tubulin on mitochondrial architecture. Mass spectrometry analysis of protein–protein interactions with β-tubulin isotypes identifies that βIII-tubulin specifically interacts with regulators of mitochondrial dynamics that may mediate these functional effects. Advanced quantitative dynamic lattice light sheet imaging of the mitochondrial network reveals that βIII-tubulin promotes a more dynamic and extended reticular mitochondrial network, and regulates mitochondrial volume. A regulatory role for the βIII-tubulin C-terminal tail in mitochondrial network dynamics and architecture has widespread implications for the maintenance of mitochondrial homeostasis in health and disease.
Collapse
|
14
|
Comandatore A, Immordino B, Balsano R, Capula M, Garajovà I, Ciccolini J, Giovannetti E, Morelli L. Potential Role of Exosomes in the Chemoresistance to Gemcitabine and Nab-Paclitaxel in Pancreatic Cancer. Diagnostics (Basel) 2022; 12:286. [PMID: 35204377 PMCID: PMC8871170 DOI: 10.3390/diagnostics12020286] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/17/2022] [Accepted: 01/20/2022] [Indexed: 02/05/2023] Open
Abstract
In recent years, a growing number of studies have evaluated the role of exosomes in pancreatic ductal adenocarcinoma cancer (PDAC) demonstrating their involvement in a multitude of pathways, including the induction of chemoresistance. The aim of this review is to present an overview of the current knowledge on the role of exosomes in the resistance to gemcitabine and nab-paclitaxel, which are two of the most commonly used drugs for the treatment of PDAC patients. Exosomes are vesicular cargos that transport multiple miRNAs, mRNAs and proteins from one cell to another cell and some of these factors can influence specific determinants of gemcitabine activity, such as the nucleoside transporter hENT1, or multidrug resistance proteins involved in the resistance to paclitaxel. Additional mechanisms underlying exosome-mediated resistance include the modulation of apoptotic pathways, cellular metabolism, or the modulation of oncogenic miRNA, such as miR-21 and miR-155. The current status of studies on circulating exosomal miRNA and their possible role as biomarkers are also discussed. Finally, we integrated the preclinical data with emerging clinical evidence, showing how the study of exosomes could help to predict the resistance of individual tumors, and guide the clinicians in the selection of innovative therapeutic strategies to overcome drug resistance.
Collapse
Affiliation(s)
- Annalisa Comandatore
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy;
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
| | - Benoit Immordino
- Fondazione Pisana per La Scienza, 56124 Pisa, Italy; (B.I.); (M.C.)
- SMARTc Unit, Centre de Recherche en Cancérologie de Marseille, Inserm U1068 Aix Marseille Université, 13385 Marseille, France;
| | - Rita Balsano
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
- Medical Oncology Unit, University Hospital of Parma, 43100 Parma, Italy;
| | - Mjriam Capula
- Fondazione Pisana per La Scienza, 56124 Pisa, Italy; (B.I.); (M.C.)
| | - Ingrid Garajovà
- Medical Oncology Unit, University Hospital of Parma, 43100 Parma, Italy;
| | - Joseph Ciccolini
- SMARTc Unit, Centre de Recherche en Cancérologie de Marseille, Inserm U1068 Aix Marseille Université, 13385 Marseille, France;
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
- Fondazione Pisana per La Scienza, 56124 Pisa, Italy; (B.I.); (M.C.)
| | - Luca Morelli
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy;
| |
Collapse
|
15
|
Won EJ, Park H, Yoon TJ, Cho YS. Gene Therapy Using Nanocarriers for Pancreatic Ductal Adenocarcinoma: Applications and Challenges in Cancer Therapeutics. Pharmaceutics 2022; 14:137. [PMID: 35057033 PMCID: PMC8780888 DOI: 10.3390/pharmaceutics14010137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/22/2021] [Accepted: 12/31/2021] [Indexed: 12/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers worldwide, and its incidence is increasing. PDAC often shows resistance to several therapeutic modalities and a higher recurrence rate after surgical treatment in the early localized stage. Combination chemotherapy in advanced pancreatic cancer has minimal impact on overall survival. RNA interference (RNAi) is a promising tool for regulating target genes to achieve sequence-specific gene silencing. Here, we summarize RNAi-based therapeutics using nanomedicine-based delivery systems that are currently being tested in clinical trials and are being developed for the treatment of PDAC. Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) genome editing has been widely used for the development of cancer models as a genetic screening tool for the identification and validation of therapeutic targets, as well as for potential cancer therapeutics. This review discusses current advances in CRISPR/Cas9 technology and its application to PDAC research. Continued progress in understanding the PDAC tumor microenvironment and nanomedicine-based gene therapy will improve the clinical outcomes of patients with PDAC.
Collapse
Affiliation(s)
- Eun-Jeong Won
- Laboratory of NanoPharmacy, College of Pharmacy, Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, 206 Worldcup-ro, Yeongtong-gu, Suwon 16499, Korea; (E.-J.W.); (T.-J.Y.)
| | - Hyeji Park
- Division of Gastroenterology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Tae-Jong Yoon
- Laboratory of NanoPharmacy, College of Pharmacy, Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, 206 Worldcup-ro, Yeongtong-gu, Suwon 16499, Korea; (E.-J.W.); (T.-J.Y.)
| | - Young-Seok Cho
- Division of Gastroenterology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| |
Collapse
|
16
|
RNAi-Based Approaches for Pancreatic Cancer Therapy. Pharmaceutics 2021; 13:pharmaceutics13101638. [PMID: 34683931 PMCID: PMC8541396 DOI: 10.3390/pharmaceutics13101638] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 01/17/2023] Open
Abstract
Pancreatic cancer is one of the most lethal forms of cancer, predicted to be the second leading cause of cancer-associated death by 2025. Despite intensive research for effective treatment strategies and novel anticancer drugs over the past decade, the overall patient survival rate remains low. RNA interference (RNAi) is capable of interfering with expression of specific genes and has emerged as a promising approach for pancreatic cancer because genetic aberrations and dysregulated signaling are the drivers for tumor formation and the stromal barrier to conventional therapy. Despite its therapeutic potential, RNA-based drugs have remaining hurdles such as poor tumor delivery and susceptibility to serum degradation, which could be overcome with the incorporation of nanocarriers for clinical applications. Here we summarize the use of small interfering RNA (siRNA) and microRNA (miRNA) in pancreatic cancer therapy in preclinical reports with approaches for targeting either the tumor or tumor microenvironment (TME) using various types of nanocarriers. In these studies, inhibition of oncogene expression and induction of a tumor suppressive response in cancer cells and surrounding immune cells in TME exhibited a strong anticancer effect in pancreatic cancer models. The review discusses the remaining challenges and prospective strategies suggesting the potential of RNAi-based therapeutics for pancreatic cancer.
Collapse
|
17
|
Conte C, Dal Poggetto G, Schiano Di Cola V, Russo A, Ungaro F, Russo G, Laurienzo P, Quaglia F. PEGylated cationic nanoassemblies based on triblock copolymers to combine siRNA therapeutics with anticancer drugs. Biomater Sci 2021; 9:6251-6265. [PMID: 34369494 DOI: 10.1039/d1bm00909e] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Nowadays, the clinical administration of siRNA therapeutics is still challenging due to the need of safe and efficient delivery carriers. In this context, biodegradable and amphiphilic triblock copolymers (ABC) containing amine-based cationic segments could be a powerful tool for siRNA delivery. Herein, we propose a range of poly(ethylene glycol) (PEG)-poly(2-dimethyl(aminoethyl) methacrylate) (pDMAEMA)-polycaprolactone (PCL) copolymers with different lengths of the blocks and hydrophilic/lipophilic balance to deliver siRNA alone or in association with a conventional anticancer drug. mPEG-pDMAEMA-PCL copolymers were synthesized by a combination of techniques and characterized by NMR analysis, Fourier transform infrared (FTIR) spectroscopy, gel permeation chromatography (GPC) and differential scanning calorimetry (DSC). Copolymers were then employed to prepare NPs through nanoprecipitation. NPs based on copolymers with long PCL chains (SSL-NPs and LLL-NPs) showed the best colloidal properties and a highly stable core-shell structure with a better orientation of the PEG fringe on the surface. Concerning siRNA delivery, SSL-NPs based on copolymers with short PEG and pDMAEMA chains showed optimized ability to complex and then deliver siRNA at the cell level. The strong interaction between the nucleic acid and the cationic pDMAEMA blocks of NPs was then confirmed by release studies that showed a sustained release of siRNA within 48 h. The transfection efficiency of NPs was assessed in human melanoma cells. NPs were complexed with a therapeutic siRNA against TUBB3 (TUB-siRNA). We observed the best results with SSL-NPs, probably due to the higher preserved buffer capacity of the pDMAEMA blocks. Finally, in order to give a proof of concept of a possible application in the combined chemo/gene-therapy of cancer, SSL-NPs complexed with TUB-siRNA were loaded with docetaxel (DTX) and then cytotoxicity was evaluated in the same cell line. The co-delivery of TUB-siRNA into NPs appeared to strongly potentiate the anti-proliferative activity of DTX, thus highlighting the combinatory activity of the NPs.
Collapse
Affiliation(s)
- Claudia Conte
- Drug Delivery Laboratory, Department of Pharmacy, University of Napoli Federico II, 80131 Napoli, Italy.
| | - Giovanni Dal Poggetto
- Institute for Polymers, Composites and Biomaterials, CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Napoli, Italy.
| | - Viola Schiano Di Cola
- Drug Delivery Laboratory, Department of Pharmacy, University of Napoli Federico II, 80131 Napoli, Italy.
| | - Annapina Russo
- Drug Delivery Laboratory, Department of Pharmacy, University of Napoli Federico II, 80131 Napoli, Italy.
| | - Francesca Ungaro
- Drug Delivery Laboratory, Department of Pharmacy, University of Napoli Federico II, 80131 Napoli, Italy.
| | - Giulia Russo
- Drug Delivery Laboratory, Department of Pharmacy, University of Napoli Federico II, 80131 Napoli, Italy.
| | - Paola Laurienzo
- Institute for Polymers, Composites and Biomaterials, CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Napoli, Italy.
| | - Fabiana Quaglia
- Drug Delivery Laboratory, Department of Pharmacy, University of Napoli Federico II, 80131 Napoli, Italy. and Institute for Polymers, Composites and Biomaterials, CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Napoli, Italy.
| |
Collapse
|
18
|
βIII-tubulin overexpression in cancer: Causes, consequences, and potential therapies. Biochim Biophys Acta Rev Cancer 2021; 1876:188607. [PMID: 34364992 DOI: 10.1016/j.bbcan.2021.188607] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/21/2021] [Accepted: 08/02/2021] [Indexed: 12/30/2022]
Abstract
Class III β-tubulin (βIII-tubulin) is frequently overexpressed in human tumors and is associated with resistance to microtubule-targeting agents, tumor aggressiveness, and poor patient outcome. Understanding the mechanisms regulating βIII-tubulin expression and the varied functions βIII-tubulin may have in different cancers is vital to assess the prognostic value of this protein and to develop strategies to enhance therapeutic benefits in βIII-tubulin overexpressing tumors. Here we gather all the available evidence regarding the clinical implications of βIII-tubulin overexpression in cancer, describe factors that regulate βIII-tubulin expression, and discuss current understanding of the mechanisms underlying βIII-tubulin-mediated resistance to microtubule-targeting agents and tumor aggressiveness. Finally, we provide an overview of emerging therapeutic strategies to target tumors that overexpress βIII-tubulin.
Collapse
|
19
|
Zhu S, Ni Y, Sun G, Wang Z, Chen J, Zhang X, Zhao J, Zhu X, Dai J, Liu Z, Liang J, Zhang H, Zhang Y, Shen P, Zeng H. Exosomal TUBB3 mRNA expression of metastatic castration-resistant prostate cancer patients: Association with patient outcome under abiraterone. Cancer Med 2021; 10:6282-6290. [PMID: 34318630 PMCID: PMC8446399 DOI: 10.1002/cam4.4168] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 02/05/2023] Open
Abstract
Background To use ddPCR to quantify plasma exosomal class III β‐tubulin (βIII‐tubulin, TUBB3, encoded by the TUBB3 gene) mRNA expression in metastatic castration‐resistant prostate cancer (mCRPC) patients, and study the association of this expression with abiraterone efficacy. Methods Blood samples were prospectively collected from 52 mCRPC patients using abiraterone as first‐line therapy to measure plasma exosomal TUBB3 mRNA expression value before the initiation of abiraterone. Study endpoints were PSA response rate, PSA‐progression‐free survival (PSA‐PFS), and overall survival (OS, from CRPC to death). Results Patients with positive exosomal TUBB3 expression showed shorter PSA‐PFS (negative TUBB3 vs. positive TUBB3: 11.0 vs. 7.9 months; p = 0.014). Further analysis demonstrated that patients with strongly positive exosomal TUBB3 (>20 copies/20 µl) was associated with even shorter PSA‐PFS (negative TUBB3 vs. positive TUBB3 [<20 copies/20 µl] vs. strongly positive TUBB3 [>20 copies/20 µl]: 11.0 vs. 8.3 vs. 3.6 months, p = 0.005). In multivariate analyzes, TUBB3 (+) (HR: 2.114, p = 0.033) and ECOG score >2 (HR: 3.039, p = 0.006) were independent prognosticators of poor PSA‐PFS. PSA response and OS did not present significant differences. Conclusion The exosomal TUBB3 mRNA expression level is associated with poor PSA‐PFS of abiraterone in mCRPC patients. The detection of exosomal TUBB3 can be valuable in their management.
Collapse
Affiliation(s)
- Sha Zhu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuchao Ni
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guangxi Sun
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zilin Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junru Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xingming Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinge Zhao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xudong Zhu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jindong Dai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhenhua Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiayu Liang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Haoran Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yaowen Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Pengfei Shen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hao Zeng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
20
|
Principe DR, Underwood PW, Korc M, Trevino JG, Munshi HG, Rana A. The Current Treatment Paradigm for Pancreatic Ductal Adenocarcinoma and Barriers to Therapeutic Efficacy. Front Oncol 2021; 11:688377. [PMID: 34336673 PMCID: PMC8319847 DOI: 10.3389/fonc.2021.688377] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/29/2021] [Indexed: 12/15/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis, with a median survival time of 10-12 months. Clinically, these poor outcomes are attributed to several factors, including late stage at the time of diagnosis impeding resectability, as well as multi-drug resistance. Despite the high prevalence of drug-resistant phenotypes, nearly all patients are offered chemotherapy leading to modest improvements in postoperative survival. However, chemotherapy is all too often associated with toxicity, and many patients elect for palliative care. In cases of inoperable disease, cytotoxic therapies are less efficacious but still carry the same risk of serious adverse effects, and clinical outcomes remain particularly poor. Here we discuss the current state of pancreatic cancer therapy, both surgical and medical, and emerging factors limiting the efficacy of both. Combined, this review highlights an unmet clinical need to improve our understanding of the mechanisms underlying the poor therapeutic responses seen in patients with PDAC, in hopes of increasing drug efficacy, extending patient survival, and improving quality of life.
Collapse
Affiliation(s)
- Daniel R. Principe
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL, United States
- Department of Surgery, University of Illinois at Chicago, Chicago, IL, United States
| | | | - Murray Korc
- Department of Developmental and Cell Biology, University of California, Irvine, CA, United States
| | - Jose G. Trevino
- Department of Surgery, Division of Surgical Oncology, Virginia Commonwealth University, Richmond, VA, United States
| | - Hidayatullah G. Munshi
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Jesse Brown VA Medical Center, Chicago, IL, United States
| | - Ajay Rana
- Department of Surgery, University of Illinois at Chicago, Chicago, IL, United States
- Jesse Brown VA Medical Center, Chicago, IL, United States
| |
Collapse
|
21
|
Sharbeen G, McCarroll JA, Akerman A, Kopecky C, Youkhana J, Kokkinos J, Holst J, Boyer C, Erkan M, Goldstein D, Timpson P, Cox TR, Pereira BA, Chitty JL, Fey SK, Najumudeen AK, Campbell AD, Sansom OJ, Ignacio RMC, Naim S, Liu J, Russia N, Lee J, Chou A, Johns A, Gill AJ, Gonzales-Aloy E, Gebski V, Guan YF, Pajic M, Turner N, Apte MV, Davis TP, Morton JP, Haghighi KS, Kasparian J, McLean BJ, Setargew YF, Phillips PA. Cancer-Associated Fibroblasts in Pancreatic Ductal Adenocarcinoma Determine Response to SLC7A11 Inhibition. Cancer Res 2021; 81:3461-3479. [PMID: 33980655 DOI: 10.1158/0008-5472.can-20-2496] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 03/01/2021] [Accepted: 04/28/2021] [Indexed: 11/16/2022]
Abstract
Cancer-associated fibroblasts (CAF) are major contributors to pancreatic ductal adenocarcinoma (PDAC) progression through protumor signaling and the generation of fibrosis, the latter of which creates a physical barrier to drugs. CAF inhibition is thus an ideal component of any therapeutic approach for PDAC. SLC7A11 is a cystine transporter that has been identified as a potential therapeutic target in PDAC cells. However, no prior study has evaluated the role of SLC7A11 in PDAC tumor stroma and its prognostic significance. Here we show that high expression of SLC7A11 in human PDAC tumor stroma, but not tumor cells, is independently prognostic of poorer overall survival. Orthogonal approaches showed that PDAC-derived CAFs are highly dependent on SLC7A11 for cystine uptake and glutathione synthesis and that SLC7A11 inhibition significantly decreases CAF proliferation, reduces their resistance to oxidative stress, and inhibits their ability to remodel collagen and support PDAC cell growth. Importantly, specific ablation of SLC7A11 from the tumor compartment of transgenic mouse PDAC tumors did not affect tumor growth, suggesting the stroma can substantially influence PDAC tumor response to SLC7A11 inhibition. In a mouse orthotopic PDAC model utilizing human PDAC cells and CAFs, stable knockdown of SLC7A11 was required in both cell types to reduce tumor growth, metastatic spread, and intratumoral fibrosis, demonstrating the importance of targeting SLC7A11 in both compartments. Finally, treatment with a nanoparticle gene-silencing drug against SLC7A11, developed by our laboratory, reduced PDAC tumor growth, incidence of metastases, CAF activation, and fibrosis in orthotopic PDAC tumors. Overall, these findings identify an important role of SLC7A11 in PDAC-derived CAFs in supporting tumor growth. SIGNIFICANCE: This study demonstrates that SLC7A11 in PDAC stromal cells is important for the tumor-promoting activity of CAFs and validates a clinically translatable nanomedicine for therapeutic SLC7A11 inhibition in PDAC.
Collapse
Affiliation(s)
- George Sharbeen
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Joshua A McCarroll
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
- Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales Sydney, New South Wales, Australia
- School of Women's and Children's Health, University of New South Wales Sydney, New South Wales, Australia
| | - Anouschka Akerman
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Chantal Kopecky
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Janet Youkhana
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - John Kokkinos
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
- Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales Sydney, New South Wales, Australia
| | - Jeff Holst
- School of Medical Science and Prince of Wales Clinical School, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Cyrille Boyer
- Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales Sydney, New South Wales, Australia
| | - Mert Erkan
- Koc University Research Centre for Translational Medicine and Department of Surgery, Koc University, School of Medicine, Istanbul, Turkey
| | - David Goldstein
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
- Prince of Wales Hospital, Prince of Wales Clinical School, Sydney, New South Wales, Australia
| | - Paul Timpson
- The Garvan Institute of Medical Research and the Kinghorn Cancer Centre, Sydney, New South Wales, Australia
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, New South Wales, Australia
- St. Vincent's Clinical School, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Thomas R Cox
- The Garvan Institute of Medical Research and the Kinghorn Cancer Centre, Sydney, New South Wales, Australia
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, New South Wales, Australia
- St. Vincent's Clinical School, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Brooke A Pereira
- The Garvan Institute of Medical Research and the Kinghorn Cancer Centre, Sydney, New South Wales, Australia
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, New South Wales, Australia
- St. Vincent's Clinical School, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Jessica L Chitty
- The Garvan Institute of Medical Research and the Kinghorn Cancer Centre, Sydney, New South Wales, Australia
- St. Vincent's Clinical School, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Sigrid K Fey
- Cancer Research UK, Beatson Institute, Glasgow, United Kingdom
| | | | | | - Owen J Sansom
- Cancer Research UK, Beatson Institute, Glasgow, United Kingdom
| | - Rosa Mistica C Ignacio
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Stephanie Naim
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Jie Liu
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Nelson Russia
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Julia Lee
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Angela Chou
- The Garvan Institute of Medical Research and the Kinghorn Cancer Centre, Sydney, New South Wales, Australia
- Department of Anatomical Pathology, Royal North Shore Hospital, University of Sydney, Sydney, New South Wales, Australia
| | - Amber Johns
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, New South Wales, Australia
| | - Anthony J Gill
- The Garvan Institute of Medical Research and the Kinghorn Cancer Centre, Sydney, New South Wales, Australia
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, New South Wales, Australia
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, Sydney, New South Wales, Australia
| | - Estrella Gonzales-Aloy
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Val Gebski
- NHMRC Clinical Trials Centre, University of Sydney, New South Wales, Australia
| | - Yi Fang Guan
- School of Medical Science and Prince of Wales Clinical School, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Marina Pajic
- The Garvan Institute of Medical Research and the Kinghorn Cancer Centre, Sydney, New South Wales, Australia
- Australian Pancreatic Cancer Genome Initiative (APGI), Sydney, New South Wales, Australia
| | - Nigel Turner
- School of Medical Sciences, University of New South Wales Sydney, New South Wales, Australia
| | - Minoti V Apte
- Pancreatic Research Group, South Western Sydney Clinical School, University New South Wales and Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Thomas P Davis
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Institute of Bioengineering & Nanotechnology, University of Queensland, Queensland, Australia
| | - Jennifer P Morton
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Koroush S Haghighi
- Prince of Wales Hospital, Prince of Wales Clinical School, Sydney, New South Wales, Australia
| | - Jorjina Kasparian
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia
| | - Benjamin J McLean
- The Garvan Institute of Medical Research and the Kinghorn Cancer Centre, Sydney, New South Wales, Australia
| | | | - Phoebe A Phillips
- Pancreatic Cancer Translational Research Group, Prince of Wales Clinical School and School of Medical Sciences, Lowy Cancer Research Centre, University of New South Wales Sydney, New South Wales, Australia.
- Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales Sydney, New South Wales, Australia
| |
Collapse
|
22
|
Waters AM, Khatib TO, Papke B, Goodwin CM, Hobbs GA, Diehl JN, Yang R, Edwards AC, Walsh KH, Sulahian R, McFarland JM, Kapner KS, Gilbert TSK, Stalnecker CA, Javaid S, Barkovskaya A, Grover KR, Hibshman PS, Blake DR, Schaefer A, Nowak KM, Klomp JE, Hayes TK, Kassner M, Tang N, Tanaseichuk O, Chen K, Zhou Y, Kalkat M, Herring LE, Graves LM, Penn LZ, Yin HH, Aguirre AJ, Hahn WC, Cox AD, Der CJ. Targeting p130Cas- and microtubule-dependent MYC regulation sensitizes pancreatic cancer to ERK MAPK inhibition. Cell Rep 2021; 35:109291. [PMID: 34192548 PMCID: PMC8340308 DOI: 10.1016/j.celrep.2021.109291] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 01/31/2021] [Accepted: 06/03/2021] [Indexed: 12/28/2022] Open
Abstract
To identify therapeutic targets for KRAS mutant pancreatic cancer, we conduct a druggable genome small interfering RNA (siRNA) screen and determine that suppression of BCAR1 sensitizes pancreatic cancer cells to ERK inhibition. Integrative analysis of genome-scale CRISPR-Cas9 screens also identify BCAR1 as a top synthetic lethal interactor with mutant KRAS. BCAR1 encodes the SRC substrate p130Cas. We determine that SRC-inhibitor-mediated suppression of p130Cas phosphorylation impairs MYC transcription through a DOCK1-RAC1-β-catenin-dependent mechanism. Additionally, genetic suppression of TUBB3, encoding the βIII-tubulin subunit of microtubules, or pharmacological inhibition of microtubule function decreases levels of MYC protein in a calpain-dependent manner and potently sensitizes pancreatic cancer cells to ERK inhibition. Accordingly, the combination of a dual SRC/tubulin inhibitor with an ERK inhibitor cooperates to reduce MYC protein and synergistically suppress the growth of KRAS mutant pancreatic cancer. Thus, we demonstrate that mechanistically diverse combinations with ERK inhibition suppress MYC to impair pancreatic cancer proliferation.
Collapse
Affiliation(s)
- Andrew M Waters
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tala O Khatib
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bjoern Papke
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Craig M Goodwin
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - G Aaron Hobbs
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - J Nathaniel Diehl
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Runying Yang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - A Cole Edwards
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Rita Sulahian
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Kevin S Kapner
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Thomas S K Gilbert
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; UNC Michael Hooker Proteomics Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Clint A Stalnecker
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sehrish Javaid
- Oral and Craniofacial Biomedicine PhD Program, School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Anna Barkovskaya
- Institute for Cancer Research, Oslo University Hospital, Oslo 0379, Norway
| | - Kajal R Grover
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Priya S Hibshman
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Devon R Blake
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Antje Schaefer
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Katherine M Nowak
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jennifer E Klomp
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tikvah K Hayes
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Michelle Kassner
- Cancer and Cell Biology Division, Translational Genomic Research Institute, Phoenix, AZ 85004, USA
| | - Nanyun Tang
- Cancer and Cell Biology Division, Translational Genomic Research Institute, Phoenix, AZ 85004, USA
| | - Olga Tanaseichuk
- Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Kaisheng Chen
- Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Yingyao Zhou
- Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Manpreet Kalkat
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5S, Canada
| | - Laura E Herring
- UNC Michael Hooker Proteomics Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lee M Graves
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Linda Z Penn
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5S, Canada
| | - Hongwei H Yin
- Cancer and Cell Biology Division, Translational Genomic Research Institute, Phoenix, AZ 85004, USA
| | - Andrew J Aguirre
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02215, USA; Brigham and Women's Hospital, Boston, MA 02215, USA
| | - William C Hahn
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02215, USA; Brigham and Women's Hospital, Boston, MA 02215, USA
| | - Adrienne D Cox
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Oral and Craniofacial Biomedicine PhD Program, School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Channing J Der
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Oral and Craniofacial Biomedicine PhD Program, School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
23
|
Albahde MAH, Abdrakhimov B, Li GQ, Zhou X, Zhou D, Xu H, Qian H, Wang W. The Role of Microtubules in Pancreatic Cancer: Therapeutic Progress. Front Oncol 2021; 11:640863. [PMID: 34094924 PMCID: PMC8176010 DOI: 10.3389/fonc.2021.640863] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 03/26/2021] [Indexed: 12/14/2022] Open
Abstract
Pancreatic cancer has an extremely low prognosis, which is attributable to its high aggressiveness, invasiveness, late diagnosis, and lack of effective therapies. Among all the drugs joining the fight against this type of cancer, microtubule-targeting agents are considered to be the most promising. They inhibit cancer cells although through different mechanisms such as blocking cell division, apoptosis induction, etc. Hereby, we review the functions of microtubule cytoskeletal proteins in tumor cells and comprehensively examine the effects of microtubule-targeting agents on pancreatic carcinoma.
Collapse
Affiliation(s)
- Mugahed Abdullah Hasan Albahde
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, China
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang University, Hangzhou, China
| | - Bulat Abdrakhimov
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guo-Qi Li
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| | - Xiaohu Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| | - Dongkai Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| | - Hao Xu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| | - Huixiao Qian
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, China
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang University, Hangzhou, China
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China
| |
Collapse
|
24
|
Hu X, Xia F, Lee J, Li F, Lu X, Zhuo X, Nie G, Ling D. Tailor-Made Nanomaterials for Diagnosis and Therapy of Pancreatic Ductal Adenocarcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002545. [PMID: 33854877 PMCID: PMC8025024 DOI: 10.1002/advs.202002545] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 10/25/2020] [Indexed: 05/05/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers worldwide due to its aggressiveness and the challenge to early diagnosis and treatment. In recent decades, nanomaterials have received increasing attention for diagnosis and therapy of PDAC. However, these designs are mainly focused on the macroscopic tumor therapeutic effect, while the crucial nano-bio interactions in the heterogeneous microenvironment of PDAC remain poorly understood. As a result, the majority of potent nanomedicines show limited performance in ameliorating PDAC in clinical translation. Therefore, exploiting the unique nature of the PDAC by detecting potential biomarkers together with a deep understanding of nano-bio interactions that occur in the tumor microenvironment is pivotal to the design of PDAC-tailored effective nanomedicine. This review will introduce tailor-made nanomaterials-enabled laboratory tests and advanced noninvasive imaging technologies for early and accurate diagnosis of PDAC. Moreover, the fabrication of a myriad of tailor-made nanomaterials for various PDAC therapeutic modalities will be reviewed. Furthermore, much preferred theranostic multifunctional nanomaterials for imaging-guided therapies of PDAC will be elaborated. Lastly, the prospects of these nanomaterials in terms of clinical translation and potential breakthroughs will be briefly discussed.
Collapse
Affiliation(s)
- Xi Hu
- Department of Clinical PharmacyZhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Researchthe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Fan Xia
- Institute of PharmaceuticsZhejiang Province Key Laboratory of Anti‐Cancer Drug ResearchHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Jiyoung Lee
- Institute of PharmaceuticsZhejiang Province Key Laboratory of Anti‐Cancer Drug ResearchHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Fangyuan Li
- Institute of PharmaceuticsZhejiang Province Key Laboratory of Anti‐Cancer Drug ResearchHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
- Key Laboratory of Biomedical Engineering of the Ministry of EducationCollege of Biomedical Engineering & Instrument ScienceZhejiang UniversityHangzhou310058China
| | - Xiaoyang Lu
- Department of Clinical PharmacyZhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Researchthe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Xiaozhen Zhuo
- Department of Cardiologythe First Affiliated HospitalXi'an Jiaotong UniversityXi'an710061China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and NanosafetyCAS Center for Excellence in NanoscienceNational Center for Nanoscience and TechnologyNo.11 Zhongguancun BeiyitiaoBeijing100190China
- GBA Research Innovation Institute for NanotechnologyGuangzhou510700China
| | - Daishun Ling
- Institute of PharmaceuticsZhejiang Province Key Laboratory of Anti‐Cancer Drug ResearchHangzhou Institute of Innovative MedicineCollege of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
- Key Laboratory of Biomedical Engineering of the Ministry of EducationCollege of Biomedical Engineering & Instrument ScienceZhejiang UniversityHangzhou310058China
| |
Collapse
|
25
|
Mosca L, Ilari A, Fazi F, Assaraf YG, Colotti G. Taxanes in cancer treatment: Activity, chemoresistance and its overcoming. Drug Resist Updat 2021; 54:100742. [PMID: 33429249 DOI: 10.1016/j.drup.2020.100742] [Citation(s) in RCA: 171] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/12/2020] [Accepted: 11/16/2020] [Indexed: 02/07/2023]
Abstract
Since 1984, when paclitaxel was approved by the FDA for the treatment of advanced ovarian carcinoma, taxanes have been widely used as microtubule-targeting antitumor agents. However, their historic classification as antimitotics does not describe all their functions. Indeed, taxanes act in a complex manner, altering multiple cellular oncogenic processes including mitosis, angiogenesis, apoptosis, inflammatory response, and ROS production. On the one hand, identification of the diverse effects of taxanes on oncogenic signaling pathways provides opportunities to apply these cytotoxic drugs in a more rational manner. On the other hand, this may facilitate the development of novel treatment modalities to surmount anticancer drug resistance. In the latter respect, chemoresistance remains a major impediment which limits the efficacy of antitumor chemotherapy. Taxanes have shown impact on key molecular mechanisms including disruption of mitotic spindle, mitosis slippage and inhibition of angiogenesis. Furthermore, there is an emerging contribution of cellular processes including autophagy, oxidative stress, epigenetic alterations and microRNAs deregulation to the acquisition of taxane resistance. Hence, these two lines of findings are currently promoting a more rational and efficacious taxane application as well as development of novel molecular strategies to enhance the efficacy of taxane-based cancer treatment while overcoming drug resistance. This review provides a general and comprehensive picture on the use of taxanes in cancer treatment. In particular, we describe the history of application of taxanes in anticancer therapeutics, the synthesis of the different drugs belonging to this class of cytotoxic compounds, their features and the differences between them. We further dissect the molecular mechanisms of action of taxanes and the molecular basis underlying the onset of taxane resistance. We further delineate the possible modalities to overcome chemoresistance to taxanes, such as increasing drug solubility, delivery and pharmacokinetics, overcoming microtubule alterations or mitotic slippage, inhibiting drug efflux pumps or drug metabolism, targeting redox metabolism, immune response, and other cellular functions.
Collapse
Affiliation(s)
- Luciana Mosca
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, P. le A. Moro 5, 00185 Rome, Italy
| | - Andrea Ilari
- Institute of Molecular Biology and Pathology, Italian National Research Council (IBPM-CNR), c/o Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy.
| | - Francesco Fazi
- Dept. Anatomical, Histological, Forensic & Orthopedic Sciences, Section of Histology and Medical Embryology, Sapienza University, Via A. Scarpa 14-16, 00161 Rome, Italy
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Lab, Faculty of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Gianni Colotti
- Institute of Molecular Biology and Pathology, Italian National Research Council (IBPM-CNR), c/o Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy.
| |
Collapse
|
26
|
Maloney SM, Hoover CA, Morejon-Lasso LV, Prosperi JR. Mechanisms of Taxane Resistance. Cancers (Basel) 2020; 12:E3323. [PMID: 33182737 PMCID: PMC7697134 DOI: 10.3390/cancers12113323] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 10/30/2020] [Accepted: 11/06/2020] [Indexed: 12/17/2022] Open
Abstract
The taxane family of chemotherapy drugs has been used to treat a variety of mostly epithelial-derived tumors and remain the first-line treatment for some cancers. Despite the improved survival time and reduction of tumor size observed in some patients, many have no response to the drugs or develop resistance over time. Taxane resistance is multi-faceted and involves multiple pathways in proliferation, apoptosis, metabolism, and the transport of foreign substances. In this review, we dive deeper into hypothesized resistance mechanisms from research during the last decade, with a focus on the cancer types that use taxanes as first-line treatment but frequently develop resistance to them. Furthermore, we will discuss current clinical inhibitors and those yet to be approved that target key pathways or proteins and aim to reverse resistance in combination with taxanes or individually. Lastly, we will highlight taxane response biomarkers, specific genes with monitored expression and correlated with response to taxanes, mentioning those currently being used and those that should be adopted. The future directions of taxanes involve more personalized approaches to treatment by tailoring drug-inhibitor combinations or alternatives depending on levels of resistance biomarkers. We hope that this review will identify gaps in knowledge surrounding taxane resistance that future research or clinical trials can overcome.
Collapse
Affiliation(s)
- Sara M. Maloney
- Harper Cancer Research Institute, South Bend, IN 46617, USA;
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, South Bend, IN 46617, USA
| | - Camden A. Hoover
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (C.A.H.); (L.V.M.-L.)
| | - Lorena V. Morejon-Lasso
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (C.A.H.); (L.V.M.-L.)
| | - Jenifer R. Prosperi
- Harper Cancer Research Institute, South Bend, IN 46617, USA;
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, South Bend, IN 46617, USA
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (C.A.H.); (L.V.M.-L.)
| |
Collapse
|
27
|
Lopes D, Maiato H. The Tubulin Code in Mitosis and Cancer. Cells 2020; 9:cells9112356. [PMID: 33114575 PMCID: PMC7692294 DOI: 10.3390/cells9112356] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/20/2020] [Accepted: 10/24/2020] [Indexed: 12/23/2022] Open
Abstract
The “tubulin code” combines different α/β-tubulin isotypes with several post-translational modifications (PTMs) to generate microtubule diversity in cells. During cell division, specific microtubule populations in the mitotic spindle are differentially modified, but only recently, the functional significance of the tubulin code, with particular emphasis on the role specified by tubulin PTMs, started to be elucidated. This is the case of α-tubulin detyrosination, which was shown to guide chromosomes during congression to the metaphase plate and allow the discrimination of mitotic errors, whose correction is required to prevent chromosomal instability—a hallmark of human cancers implicated in tumor evolution and metastasis. Although alterations in the expression of certain tubulin isotypes and associated PTMs have been reported in human cancers, it remains unclear whether and how the tubulin code has any functional implications for cancer cell properties. Here, we review the role of the tubulin code in chromosome segregation during mitosis and how it impacts cancer cell properties. In this context, we discuss the existence of an emerging “cancer tubulin code” and the respective implications for diagnostic, prognostic and therapeutic purposes.
Collapse
Affiliation(s)
- Danilo Lopes
- Chromosome Instability & Dynamics Group, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal;
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Helder Maiato
- Chromosome Instability & Dynamics Group, i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal;
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Cell Division Group, Experimental Biology Unit, Department of Biomedicine, Faculdade de Medicina, Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- Correspondence: ; Tel.: +351-22-040-8800
| |
Collapse
|
28
|
Borys F, Joachimiak E, Krawczyk H, Fabczak H. Intrinsic and Extrinsic Factors Affecting Microtubule Dynamics in Normal and Cancer Cells. Molecules 2020; 25:E3705. [PMID: 32823874 PMCID: PMC7464520 DOI: 10.3390/molecules25163705] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/03/2020] [Accepted: 08/08/2020] [Indexed: 12/18/2022] Open
Abstract
Microtubules (MTs), highly dynamic structures composed of α- and β-tubulin heterodimers, are involved in cell movement and intracellular traffic and are essential for cell division. Within the cell, MTs are not uniform as they can be composed of different tubulin isotypes that are post-translationally modified and interact with different microtubule-associated proteins (MAPs). These diverse intrinsic factors influence the dynamics of MTs. Extrinsic factors such as microtubule-targeting agents (MTAs) can also affect MT dynamics. MTAs can be divided into two main categories: microtubule-stabilizing agents (MSAs) and microtubule-destabilizing agents (MDAs). Thus, the MT skeleton is an important target for anticancer therapy. This review discusses factors that determine the microtubule dynamics in normal and cancer cells and describes microtubule-MTA interactions, highlighting the importance of tubulin isoform diversity and post-translational modifications in MTA responses and the consequences of such a phenomenon, including drug resistance development.
Collapse
Affiliation(s)
- Filip Borys
- Laboratory of Cytoskeleton and Cilia Biology Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland;
- Department of Organic Chemistry, Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego Street, 00-664 Warsaw, Poland;
| | - Ewa Joachimiak
- Laboratory of Cytoskeleton and Cilia Biology Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland;
| | - Hanna Krawczyk
- Department of Organic Chemistry, Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego Street, 00-664 Warsaw, Poland;
| | - Hanna Fabczak
- Laboratory of Cytoskeleton and Cilia Biology Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland;
| |
Collapse
|
29
|
Kamal MA, Al-Zahrani MH, Khan SH, Khan MH, Al-Subhi HA, Kuerban A, Aslam M, Al-Abbasi FA, Anwar F. Tubulin Proteins in Cancer Resistance: A Review. Curr Drug Metab 2020; 21:178-185. [DOI: 10.2174/1389200221666200226123638] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 11/26/2019] [Accepted: 12/03/2019] [Indexed: 12/26/2022]
Abstract
Cancer cells are altered with cell cycle genes or they are mutated, leading to a high rate of proliferation
compared to normal cells. Alteration in these genes leads to mitosis dysregulation and becomes the basis of tumor
progression and resistance to many drugs. The drugs which act on the cell cycle fail to arrest the process, making
cancer cell non-responsive to apoptosis or cell death. Vinca alkaloids and taxanes fall in this category and are
referred to as antimitotic agents. Microtubule proteins play an important role in mitosis during cell division as a
target site for vinca alkaloids and taxanes. These proteins are dynamic in nature and are composed of α-β-tubulin
heterodimers. β-tubulin specially βΙΙΙ isotype is generally altered in expression within cancerous cells. Initially,
these drugs were very effective in the treatment of cancer but failed to show their desired action after initial
chemotherapy. The present review highlights some of the important targets and their mechanism of resistance
offered by cancer cells with new promising drugs from natural sources that can lead to the development of a new
approach to chemotherapy.
Collapse
Affiliation(s)
- Mohammad Amjad Kamal
- Metabolomics and Enzymology Unit, Fundamental and Applied Biology Group, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Maryam Hassan Al-Zahrani
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Salman Hasan Khan
- Department of Orthodontics, and Dentofacial Orthopaedics, TMU Dental College, Moradabad, Uttar Pradesh, India
| | - Mateen Hasan Khan
- Department of Pharmacology, Shri Venkateshwara University, Gajraula, Amroha, Uttar Pradesh, India
| | - Hani Awad Al-Subhi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Abudukadeer Kuerban
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Muhammad Aslam
- Department of Statistics, Faculy of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Fahad Ahmed Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Firoz Anwar
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
30
|
Liu Q, Cai P, Guo S, Shi J, Sun H. Identification of a lathyrane-type diterpenoid EM-E-11-4 as a novel paclitaxel resistance reversing agent with multiple mechanisms of action. Aging (Albany NY) 2020; 12:3713-3729. [PMID: 32108588 PMCID: PMC7066893 DOI: 10.18632/aging.102842] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 02/04/2020] [Indexed: 12/17/2022]
Abstract
P-glycoprotein (P-gp) and βIII-tubulin overexpression-mediated drug resistance leads to clinical therapy failure for paclitaxel. However, the development of paclitaxel-resistance reversal agents has not had much success. In this study, EM-E-11-4, a lathyrane-type diterpenoid extracted from Euphorbia micractina, demonstrated good anti-MDR (multidrug resistance) activity in paclitaxel-resistant tumor cells overexpressing either P-gp or βIII-tubulin. EM-E-11-4 was able to recover the effects of paclitaxel in inducing arrest at G2/M phase and apoptosis in both A549/Tax (P-gp overexpression) and Hela/βIII (βIII-tubulin overexpression) cells, respectively, at a non-cytotoxic dose. EM-E-11-4 could enable Flutax-1 and Rhodamine 123 be accumulated intracellularly at an accelerating rate in A549/Tax cells by inhibiting the activity of P-gp ATPase, rather than affecting the expression of P-gp. In addition, it also strengthened the effects of paclitaxel in promoting tubulin polymerization and the binding of paclitaxel to microtubules in vitro. It inhibited the expression of βIII-tubulin in Hela/βIII cells in a dose-dependent manner while not exerting influence on the other β-tubulin subtypes. As far as we know, this is the first study to report that a small molecule natural product could specifically inhibit the expression of βIII-tubulin. These results suggest EM-E-11-4 may serve as a promising MDR reversal agent, particularly for patients bearing tumors with high expression of P-gp and βIII-tubulin.
Collapse
Affiliation(s)
- Qian Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.,Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Pei Cai
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha 410008, China
| | - Siwei Guo
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Jiangong Shi
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Hua Sun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
31
|
Tubulin heterogeneity regulates functions and dynamics of microtubules and plays a role in the development of drug resistance in cancer. Biochem J 2019; 476:1359-1376. [DOI: 10.1042/bcj20190123] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/21/2019] [Accepted: 04/24/2019] [Indexed: 01/01/2023]
Abstract
Abstract
Microtubules, composed of αβ-tubulin heterodimers, exhibit diverse structural and functional properties in different cell types. The diversity in the microtubule structure originates from tubulin heterogeneities, namely tubulin isotypes and their post-translational modifications (PTMs). These heterogeneities confer differential stability to microtubules and provide spatial cues for the functioning of the cell. Furthermore, the altered expressions of tubulin isotypes and PTMs are prominent factors for the development of resistance against some cancer drugs. In this review, we summarize our current knowledge of the tubulin isotypes and PTMs and how, together, they control the cellular functions of the microtubules. We also describe how cancer cells use this tubulin heterogeneity to acquire resistance against clinical agents and discuss existing attempts to counter the developed resistance.
Collapse
|
32
|
Zavala WD, Foscolo MR, Kunda PE, Cavicchia JC, Acosta CG. Changes in the expression of the potassium channels TASK1, TASK3 and TRESK in a rat model of oral squamous cell carcinoma and their relation to malignancy. Arch Oral Biol 2019; 100:75-85. [PMID: 30818127 DOI: 10.1016/j.archoralbio.2019.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/30/2019] [Accepted: 02/15/2019] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Potassium channels have been proposed to promote cancer cell proliferation and metastases. Thus, we investigated the expression pattern of three 2-pore domain potassium channels (K2Ps) TASK1, TASK3 and TRESK in advanced oral squamous cell carcinoma (OSCC), the commonest oral malignancy. DESIGN We used 4-nitroquinoline-1-oxide (4-NQO) to induce high grade OSCC in male adult rats. We then used immunohistochemistry and Western blotting to study the distribution and expression pattern of TASK1, TASK3 and TRESK in normal versus cancerous tissue. We also examined the expression of β-tubulin III (β-tub3), a marker associated with resistance to taxane-based chemotherapy and poor patient prognosis, and its correlation with the K2Ps. Finally, we studied the expression of TASK1, TASK3 and TRESK in human samples of SCC of oral origin. RESULTS We found that TASK3 was significantly up-regulated whereas TASK1 and TRESK were both significantly down-regulated in advanced, poorly differentiated OSCC. Both, rat and human SCC showed a significant increase in the expression of β-tub3. Interestingly, the expression of the latter correlated positively and significantly with TASK3 and TRESK but not TASK1 in rat OSCC. Our initial results showed a similar pattern of up and down regulation and correlation with β-tub3 for these three K2Ps in human SCC. CONCLUSIONS The changes in expression and the co-localization with a marker of resistance to taxanes like β-tub3 turn TASK1, TASK3 and TRESK into potentially new prognostic tools and possibly new therapeutic targets for OSCC.
Collapse
Affiliation(s)
- Walther D Zavala
- Facultad de Odontología, Universidad Nacional de Cuyo, Mendoza, Argentina.
| | - Mabel R Foscolo
- Instituto de Histología y Embriología de Mendoza "Dr. M. Burgos" (IHEM-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.
| | - Patricia E Kunda
- Centro Investigación Medicina Traslacional "Severo Amuchástegui" (CIMETSA), Instituto Universitario Ciencias Biomédicas Córdoba (IUCBC), Córdoba, Argentina.
| | - Juan C Cavicchia
- Instituto de Histología y Embriología de Mendoza "Dr. M. Burgos" (IHEM-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.
| | - Cristian G Acosta
- Instituto de Histología y Embriología de Mendoza "Dr. M. Burgos" (IHEM-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
33
|
Kashyap VK, Wang Q, Setua S, Nagesh PKB, Chauhan N, Kumari S, Chowdhury P, Miller DD, Yallapu MM, Li W, Jaggi M, Hafeez BB, Chauhan SC. Therapeutic efficacy of a novel βIII/βIV-tubulin inhibitor (VERU-111) in pancreatic cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:29. [PMID: 30674344 PMCID: PMC6343279 DOI: 10.1186/s13046-018-1009-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/17/2018] [Indexed: 12/15/2022]
Abstract
Background The management of pancreatic cancer (PanCa) is exceptionally difficult due to poor response to available therapeutic modalities. Tubulins play a major role in cell dynamics, thus are important molecular targets for cancer therapy. Among various tubulins, βIII and βIV-tubulin isoforms have been primarily implicated in PanCa progression, metastasis and chemo-resistance. However, specific inhibitors of these isoforms that have potent anti-cancer activity with low toxicity are not readily available. Methods We determined anti-cancer molecular mechanisms and therapeutic efficacy of a novel small molecule inhibitor (VERU-111) using in vitro (MTS, wound healing, Boyden chamber and real-time xCELLigence assays) and in vivo (xenograft studies) models of PanCa. The effects of VERU-111 treatment on the expression of β-tubulin isoforms, apoptosis, cancer markers and microRNAs were determined by Western blot, immunohistochemistry (IHC), confocal microscopy, qRT-PCR and in situ hybridization (ISH) analyses. Results We have identified a novel small molecule inhibitor (VERU-111), which preferentially represses clinically important, βIII and βIV tubulin isoforms via restoring the expression of miR-200c. As a result, VERU-111 efficiently inhibited tumorigenic and metastatic characteristics of PanCa cells. VERU-111 arrested the cell cycle in the G2/M phase and induced apoptosis in PanCa cell lines via modulation of cell cycle regulatory (Cdc2, Cdc25c, and Cyclin B1) and apoptosis - associated (Bax, Bad, Bcl-2, and Bcl-xl) proteins. VERU-111 treatment also inhibited tumor growth (P < 0.01) in a PanCa xenograft mouse model. Conclusions This study has identified an inhibitor of βIII/βIV tubulins, which appears to have excellent potential as monotherapy or in combination with conventional therapeutic regimens for PanCa treatment. Electronic supplementary material The online version of this article (10.1186/s13046-018-1009-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vivek K Kashyap
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Qinghui Wang
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Saini Setua
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Prashanth K B Nagesh
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Neeraj Chauhan
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Sonam Kumari
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Pallabita Chowdhury
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Duane D Miller
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Murali M Yallapu
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Wei Li
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Meena Jaggi
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA.
| | - Bilal Bin Hafeez
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA.
| | - Subhash C Chauhan
- Department of Pharmaceutical Sciences, Institute of Biomarker and Molecular Therapeutics (IBMT), College of Pharmacy, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA.
| |
Collapse
|
34
|
McCarroll JA, Sharbeen G, Kavallaris M, Phillips PA. The Use of Star Polymer Nanoparticles for the Delivery of siRNA to Mouse Orthotopic Pancreatic Tumor Models. Methods Mol Biol 2019; 1974:329-353. [PMID: 31099013 DOI: 10.1007/978-1-4939-9220-1_23] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Pancreatic cancer is a lethal malignancy which is refractory to most chemotherapy drugs. Recent landmark studies have shed new light on the complex genetic heterogeneity of pancreatic cancer and provide an opportunity to utilize "precision-based medicines" to target genes based on the genetic profile of an individual's tumor to increase the efficiency of chemotherapy and decrease tumor growth and metastases. Gene-silencing drugs in the form of short-interfering RNA (siRNA) have the potential to play an important role in precision medicine for pancreatic cancer by silencing the expression of genes including those considered difficult to inhibit (undruggable) using chemical agents. However, before siRNA can reach its clinical potential a delivery vehicle is needed to carry siRNA across the cell membrane and into the cytoplasm of the cell. Herein, we detail the methods required to use star polymer nanoparticles to deliver siRNA to pancreatic tumors in an orthotopic pancreatic cancer mouse model to silence the expression of an "undruggable" gene (βIII-tubulin) that regulates pancreatic cancer growth and chemosensitivity.
Collapse
Affiliation(s)
- Joshua A McCarroll
- Tumour Biology and Targeting Program, Lowy Cancer Research Centre, Children's Cancer Institute, UNSW Sydney, Sydney, NSW, Australia.,Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, UNSW Sydney, Sydney, NSW, Australia
| | - George Sharbeen
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Maria Kavallaris
- Tumour Biology and Targeting Program, Lowy Cancer Research Centre, Children's Cancer Institute, UNSW Sydney, Sydney, NSW, Australia.,Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, UNSW Sydney, Sydney, NSW, Australia
| | - Phoebe A Phillips
- Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, UNSW Sydney, Sydney, NSW, Australia. .,Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
35
|
TAPping into the treasures of tubulin using novel protein production methods. Essays Biochem 2018; 62:781-792. [PMID: 30429282 PMCID: PMC6281476 DOI: 10.1042/ebc20180033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/03/2018] [Accepted: 10/22/2018] [Indexed: 01/02/2023]
Abstract
Microtubules are cytoskeletal elements with important cellular functions, whose dynamic behaviour and properties are in part regulated by microtubule-associated proteins (MAPs). The building block of microtubules is tubulin, a heterodimer of α- and β-tubulin subunits. Longitudinal interactions between tubulin dimers facilitate a head-to-tail arrangement of dimers into protofilaments, while lateral interactions allow the formation of a hollow microtubule tube that mostly contains 13 protofilaments. Highly homologous α- and β-tubulin isotypes exist, which are encoded by multi-gene families. In vitro studies on microtubules and MAPs have largely relied on brain-derived tubulin preparations. However, these consist of an unknown mix of tubulin isotypes with undefined post-translational modifications. This has blocked studies on the functions of tubulin isotypes and the effects of tubulin mutations found in human neurological disorders. Fortunately, various methodologies to produce recombinant mammalian tubulins have become available in the last years, allowing researchers to overcome this barrier. In addition, affinity-based purification of tagged tubulins and identification of tubulin-associated proteins (TAPs) by mass spectrometry has revealed the 'tubulome' of mammalian cells. Future experiments with recombinant tubulins should allow a detailed description of how tubulin isotype influences basic microtubule behaviour, and how MAPs and TAPs impinge on tubulin isotypes and microtubule-based processes in different cell types.
Collapse
|
36
|
Klepinin A, Ounpuu L, Mado K, Truu L, Chekulayev V, Puurand M, Shevchuk I, Tepp K, Planken A, Kaambre T. The complexity of mitochondrial outer membrane permeability and VDAC regulation by associated proteins. J Bioenerg Biomembr 2018; 50:339-354. [PMID: 29998379 PMCID: PMC6209068 DOI: 10.1007/s10863-018-9765-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 07/05/2018] [Indexed: 12/18/2022]
Abstract
Previous studies have shown that class II β-tubulin plays a key role in the regulation of oxidative phosphorylation (OXPHOS) in some highly differentiated cells, but its role in malignant cells has remained unclear. To clarify these aspects, we compared the bioenergetic properties of HL-1 murine sarcoma cells, murine neuroblastoma cells (uN2a) and retinoic acid - differentiated N2a cells (dN2a). We examined the expression and possible co-localization of mitochondrial voltage dependent anion channel (VDAC) with hexokinase-2 (HK-2) and βII-tubulin, the role of depolymerized βII-tubuline and the effect of both proteins in the regulation of mitochondrial outer membrane (MOM) permeability. Our data demonstrate that neuroblastoma and sarcoma cells are prone to aerobic glycolysis, which is partially mediated by the presence of VDAC bound HK-2. Microtubule destabilizing (colchicine) and stabilizing (taxol) agents do not affect the MOM permeability for ADP in N2a and HL-1 cells. The obtained results show that βII-tubulin does not regulate the MOM permeability for adenine nucleotides in these cells. HL-1 and NB cells display comparable rates of ADP-activated respiration. It was also found that differentiation enhances the involvement of OXPHOS in N2a cells due to the rise in their mitochondrial reserve capacity. Our data support the view that the alteration of mitochondrial affinity for ADNs is one of the characteristic features of cancer cells. It can be concluded that the binding sites for tubulin and hexokinase within the large intermembrane protein supercomplex Mitochondrial Interactosome, could be different between muscle and cancer cells.
Collapse
Affiliation(s)
- Aleksandr Klepinin
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618, Tallinn, Estonia
| | - Lyudmila Ounpuu
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618, Tallinn, Estonia
| | - Kati Mado
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618, Tallinn, Estonia
| | - Laura Truu
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618, Tallinn, Estonia
| | - Vladimir Chekulayev
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618, Tallinn, Estonia
| | - Marju Puurand
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618, Tallinn, Estonia
| | - Igor Shevchuk
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618, Tallinn, Estonia
| | - Kersti Tepp
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618, Tallinn, Estonia
| | - Anu Planken
- Oncology and Hematology Clinic at the North Estonia Medical Centre, Tallinn, Estonia
| | - Tuuli Kaambre
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618, Tallinn, Estonia.
| |
Collapse
|
37
|
Kato A, Naiki-Ito A, Naitoh I, Hayashi K, Nakazawa T, Shimizu S, Nishi Y, Okumura F, Inoue T, Takada H, Kondo H, Yoshida M, Takahashi S, Joh T. The absence of class III β-tubulin is predictive of a favorable response to nab-paclitaxel and gemcitabine in patients with unresectable pancreatic ductal adenocarcinoma. Hum Pathol 2018; 74:92-98. [DOI: 10.1016/j.humpath.2018.01.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 01/04/2018] [Accepted: 01/05/2018] [Indexed: 11/17/2022]
|
38
|
Marine Bacterial Polysaccharide EPS11 Inhibits Cancer Cell Growth via Blocking Cell Adhesion and Stimulating Anoikis. Mar Drugs 2018. [PMID: 29518055 PMCID: PMC5867629 DOI: 10.3390/md16030085] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Tumor cells that acquire metastatic potential have developed resistance to anoikis, a cell death process, after detachment from their primary site to the second organ. In this study, we investigated the molecular mechanisms of a novel marine bacterial polysaccharide EPS11 which exerts its cytotoxic effects through affecting cancer cell adhesion and anoikis. Firstly, we found that EPS11 could significantly affect cell proliferation and block cell adhesion in A549 cells. We further demonstrated that the expression of several cell adhesion associated proteins is downregulated and the filiform structures of cancer cells are destroyed after EPS11 treatment. Interestingly, the destruction of filiform structures in A549 cells by EPS11 is in a dose-dependent manner, and the inhibitory tendency is very consistent with that observed in the cell adhesion assay, which confirms that filiform structures play important roles in modulating cell adhesion. Moreover, we showed that EPS11 induces apoptosis of A549 cells through stimulating βIII-tubulin associated anoikis: (i) EPS11 inhibits the expression of βIII-tubulin in both transcription and translation levels; and (ii) EPS11 treatment dramatically decreases the phosphorylation of protein kinase B (PKB or AKT), a critical downstream effector of βIII-tubulin. Importantly, EPS11 evidently inhibits the growth of A549-derived tumor xenografts in vivo. Thus, our results suggest that EPS11 may be a potential candidate for human non-small cell lung carcinoma treatment via blocking filiform structure mediated adhesion and stimulating βIII-tubulin associated anoikis.
Collapse
|
39
|
Ferguson SD, Xiu J, Weathers SP, Zhou S, Kesari S, Weiss SE, Verhaak RG, Hohl RJ, Barger GR, Reddy SK, Heimberger AB. GBM-associated mutations and altered protein expression are more common in young patients. Oncotarget 2018; 7:69466-69478. [PMID: 27579614 PMCID: PMC5342491 DOI: 10.18632/oncotarget.11617] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 08/15/2016] [Indexed: 12/16/2022] Open
Abstract
Background Geriatric glioblastoma (GBM) patients have a poorer prognosis than younger patients, but IDH1/2 mutations (more common in younger patients) confer a favorable prognosis. We compared key GBM molecular alterations between an elderly (age ≥ 70) and younger (18 < = age < = 45) cohort to explore potential therapeutic opportunities. Results Alterations more prevalent in the young GBM cohort compared to the older cohort (P < 0.05) were: overexpression of ALK, RRM1, TUBB3 and mutation of ATRX, BRAF, IDH1, and TP53. However, PTEN mutation was significantly more frequent in older patients. Among patients with wild-type IDH1/2 status, TOPO1 expression was higher in younger patients, whereas MGMT methylation was more frequent in older patients. Within the molecularly-defined IDH wild-type GBM cohort, younger patients had significantly more mutations in PDGFRA, PTPN11, SMARCA4, BRAF and TP53. Methods GBMs from 178 elderly patients and 197 young patients were analyzed using DNA sequencing, immunohistochemistry, in situ hybridization, and MGMT-methylation assay to ascertain mutational and amplification/expressional status. Conclusions Significant molecular differences occurred in GBMs from elderly and young patients. Except for the older cohort's more frequent PTEN mutation and MGMT methylation, younger patients had a higher frequency of potential therapeutic targets.
Collapse
Affiliation(s)
- Sherise D Ferguson
- Departments of Neurosurgery, Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Joanne Xiu
- Caris Life Sciences, Phoenix, AZ 85040, USA
| | - Shiao-Pei Weathers
- Departments of Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Shouhao Zhou
- Departments of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Santosh Kesari
- Department of Translational Neuro-Oncology and Neurotherapeutics, Pacific Neuroscience Institute and John Wayne Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA 90404, USA
| | | | - Roeland G Verhaak
- Department of Genome Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77054, USA
| | - Raymond J Hohl
- Penn State Hershey Cancer Institute, Hershey, PA 17033, USA
| | - Geoffrey R Barger
- Department of Neurology, Wayne State University, School of Medicine, Karmanos Cancer Center, Detroit, MI 48201, USA
| | | | - Amy B Heimberger
- Departments of Neurosurgery, Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
40
|
Ferguson SD, Zhou S, Xiu J, Hashimoto Y, Sanai N, Kim L, Kesari S, de Groot J, Spetzler D, Heimberger AB. Ependymomas overexpress chemoresistance and DNA repair-related proteins. Oncotarget 2018; 9:7822-7831. [PMID: 29487694 PMCID: PMC5814261 DOI: 10.18632/oncotarget.23288] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/05/2017] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND After surgery and radiation, treatment options for ependymoma are few making recurrence a challenging issue. Specifically, the efficacy of chemotherapy at recurrence is limited. We performed molecular profiling on a cohort of ependymoma cases in order to uncover therapeutic targets and to elucidate the molecular mechanisms contributing to treatment resistance. RESULTS This ependymoma cohort showed minimal alterations in gene amplifications and mutations but had high expression rates of DNA synthesis and repair enzymes such as RRM1 (47%), ERCC1 (48%), TOPO1 (62%) and class III β-tublin (TUBB3) (57%), which are also all associated with chemoresistance. This cohort also had high expression rates of transporter proteins that mediate multi-drug resistance including BCRP (71%) and MRP1 (43%). Subgroup analyses showed that cranial ependymomas expressed the DNA synthesis enzyme TS significantly more frequently than spinal lesions did (57% versus 15%; p = 0.0328) and that increased TS expression was correlated with increased tumor grade (p = 0.0009). High-grade lesions were also significantly associated with elevated expression of TOP2A (p = 0.0092) and TUBB3 (p = 0.0157). MATERIALS AND METHODS We reviewed the characteristics of 41 ependymomas (21 cranial, 20 spinal; 8 grade I, 11 grade II, 22 grade III) that underwent multiplatform profiling with immunohistochemistry, next-generation sequencing, and in situ hybridization. CONCLUSIONS Ependymomas are enriched with proteins involved in chemoresistance and in DNA synthesis and repair, which is consistent with the meager clinical effectiveness of conventional systemic therapy in ependymoma. Adjuvant therapies that combine conventional chemotherapy with the inhibition of chemoresistance-related proteins may represent a novel treatment paradigm for this difficult disease.
Collapse
Affiliation(s)
- Sherise D. Ferguson
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shouhao Zhou
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Yuuri Hashimoto
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nader Sanai
- Division of Neurosurgical Oncology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Lyndon Kim
- Department of Neurological Surgery and Medical Oncology, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Santosh Kesari
- Department of Translational Neurosciences and Neurotherapeutics, Pacific Neuroscience Institute and John Wayne Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, USA
| | - John de Groot
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Spetzler
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amy B. Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
41
|
Lin C, Zhao GC, Xu YD, Wang DS, Jin DY, Ji Y, Lou WH, Wu WC. Increased expression of αTubulin is associated with poor prognosis in patients with pancreatic cancer after surgical resection. Oncotarget 2018; 7:60657-60664. [PMID: 27447976 PMCID: PMC5312409 DOI: 10.18632/oncotarget.10630] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 06/07/2016] [Indexed: 12/17/2022] Open
Abstract
Background αTubulin, the essential orchestrator of cytoskeletal protein polymers, critical for cell growth and division, motility, signaling development and maintenance of cell shape, plays vital roles in the oncogenesis and progression of various types of cancer, but its role in prognosis of pancreatic cancer patients remains unknown. The aim of this study was to investigate its prognostic value in patients with pancreatic cancer after surgical resection. Results αTubulin expression in pancreatic cancer was significantly associated with N classification (p = 0.013) and TNM stage (p = 0.025). Increased expression of αTubulin in tumoral tissue was associated with decreased overall survival rate (p = 0.002). Multivariate Cox regression analysis suggested that αTubulin expression was an independent prognostic indicator for pancreatic cancer except for T and N classification (p = 0.002). Using multivariate analysis, αTubulin expression, CA19-9, and N classification were selected to generate the nomogram to predict the 1-year and 3-year overall survival. The c-index of this model was 0.692. The calibration curve for probability of survival showed good agreement between prediction by nomogram and actual observation. Methods αTubulin expression was evaluated by tissue microarrays from 124 pancreatic cancer patients and statistically assessed for correlations with the clinical profiles and the prognosis of the patients with pancreatic cancer. The prognostic nomogram was designed to predict 1-year and 3-year overall survival probability. Conclusions αTubulin expression might be an independent prognostic factor for pancreatic cancer after surgical resection and could potentially be a high-priority therapeutic target. Incorporating αTubulin expression into CA19-9 and N classification can provide a good prognostic model.
Collapse
Affiliation(s)
- Chao Lin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guo-Chao Zhao
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ya-Dong Xu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dan-Song Wang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Da-Yong Jin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuan Ji
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wen-Hui Lou
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wen-Chuan Wu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
42
|
Tame MA, Manjón AG, Belokhvostova D, Raaijmakers JA, Medema RH. TUBB3 overexpression has a negligible effect on the sensitivity to taxol in cultured cell lines. Oncotarget 2017; 8:71536-71547. [PMID: 29069726 PMCID: PMC5641069 DOI: 10.18632/oncotarget.17740] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 04/22/2017] [Indexed: 12/11/2022] Open
Abstract
Microtubules are cellular targets for a variety of anticancer therapies because of their critical function in mitosis. Taxol belongs to a class of microtubule targeting agents that suppresses microtubule dynamics and interferes with the functioning of the mitotic spindle, thereby effectively blocking cell cycle progression of rapidly proliferating tumor cells. Despite its antitumor activity, drug resistance remains a common obstacle in improving its overall clinical efficacy. Previous studies have shown that the expression of a specific β-tubulin isotype, βIII-tubulin/TUBB3, is dysregulated in drug-refractory tumors. However, whether enhanced TUBB3 expression is directly involved in promoting taxol resistance remains a subject of debate. Here, we have used several approaches to assess the functional relation of TUBB3 overexpression and taxol resistance. First, we generated a number of taxol-resistant cell lines, to find that TUBB3 expression was elevated in a resistant cell line (RPE-20) derived from untransformed retinal pigment epithelial (RPE) cells, but the abundance of TUBB3 remained unchanged in four other cell lines after taxol treatment. However, although RPE-20 cells displayed enhanced TUBB3 levels, we find that simultaneous up-regulation of the P-glycoprotein (P-gP) drug-efflux pump is responsible for the resistance to taxol. Indeed, we could show that TUBB3 levels were dynamically regulated upon taxol exposure and withdrawal, unrelated to the resistance phenotype. Next, we generated cell lines in which we could induce robust overexpression of TUBB3 from its endogenous locus employing the CRISPRa system. We demonstrate that solely enhancing TUBB3 expression results in a very minor decrease in the sensitivity to taxol. This was further substantiated by selective depletion of TUBB3 in a series of breast cancer cell lines expressing high levels of TUBB3. We find that TUBB3 depletion had a minimal effect on the sensitivity to taxol in one of these cell lines, but had no effect in all of the others. Based on these findings we propose that TUBB3 overexpression can only marginally affect the sensitivity to taxol in cultured cell lines.
Collapse
Affiliation(s)
- Mihoko A. Tame
- Division of Cell Biology and Cancer Genomics Center, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Anna G. Manjón
- Division of Cell Biology and Cancer Genomics Center, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Daria Belokhvostova
- Division of Cell Biology and Cancer Genomics Center, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Jonne A. Raaijmakers
- Division of Cell Biology and Cancer Genomics Center, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - René H. Medema
- Division of Cell Biology and Cancer Genomics Center, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
43
|
Tóth C, Sükösd F, Valicsek E, Herpel E, Schirmacher P, Renner M, Mader C, Tiszlavicz L, Kriegsmann J. Expression of ERCC1, RRM1, TUBB3 in correlation with apoptosis repressor ARC, DNA mismatch repair proteins and p53 in liver metastasis of colorectal cancer. Int J Mol Med 2017; 40:1457-1465. [PMID: 28949378 PMCID: PMC5627886 DOI: 10.3892/ijmm.2017.3136] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 08/25/2017] [Indexed: 12/14/2022] Open
Abstract
Liver metastasis in colorectal cancer is common and the primary treatment is chemotherapy. To date, there is no routinely used test in clinical practice to predict the effectiveness of conventional chemotherapy. Therefore, biomarkers with predictive value for conventional chemotherapy would be of considerable benefit in treatment planning. We analysed three proteins [excision repair cross-complementing 1 (ERCC1), ribonucleoside-diphosphate reductase 1 (RRM1) and class III β-tubulin (TUBB3)] in colorectal cancer liver metastasis. We used tissue microarray slides with 101 liver metastasis samples, stained for ERCC1, RRM1 and TUBB3 and established scoring systems (fitted for tissue microarray) for each protein. In statistical analysis, we compared the expression of ERCC1, RRM1 and TUBB3 to mismatch proteins (MLH1, MSH2, MSH6 and PMS2), p53 and to apoptosis repressor protein (ARC). Statistically significant correlations were found between ERCC1, TUBB3 and MLH1, MSH2 and RRM1 and MSH2, MSH6. Noteworthy, our analysis revealed a strong significant correlation between cytoplasmic ARC expression and RRM1, TUBB3 (p=0.000 and p=0.001, respectively), implying an additional role of TUBB3 and RRM1 not only in therapy resistance, but also in the apoptotic machinery. Our data strengthens the importance of ERCC1, TUBB3 and RRM1 in the prediction of chemotherapy effectiveness and suggest new functional connections in DNA repair, microtubule network and apoptotic signaling (i.e. ARC protein). In conclusion, we showed the importance and need of predictive biomarkers in metastasized colorectal cancer and pointed out the relevance not only of single predictive markers but also of their interactions with other known and newly explored relations between different signaling pathways.
Collapse
Affiliation(s)
- Csaba Tóth
- Institute of Pathology, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Farkas Sükösd
- Department of Pathology, University of Szeged, H-6720 Szeged, Hungary
| | - Erzsébet Valicsek
- Department of Oncotherapy, University of Szeged, H-6720 Szeged, Hungary
| | - Esther Herpel
- Institute of Pathology, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Peter Schirmacher
- Institute of Pathology, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Marcus Renner
- Institute of Pathology, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Christoph Mader
- Institute of Pathology, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - László Tiszlavicz
- Department of Pathology, University of Szeged, H-6720 Szeged, Hungary
| | - Jörg Kriegsmann
- MVZ for Histology, Cytology and Molecular Diagnostics, Trier, D-54296 Trier, Germany
| |
Collapse
|
44
|
Mitotic cell death induction by targeting the mitotic spindle with tubulin-inhibitory indole derivative molecules. Oncotarget 2017; 8:19738-19759. [PMID: 28160569 PMCID: PMC5386718 DOI: 10.18632/oncotarget.14980] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 01/06/2017] [Indexed: 01/08/2023] Open
Abstract
Tubulin-targeting molecules are widely used cancer therapeutic agents. They inhibit microtubule-based structures, including the mitotic spindle, ultimately preventing cell division. The final fates of microtubule-inhibited cells are however often heterogeneous and difficult to predict. While recent work has provided insight into the cell response to inhibitors of microtubule dynamics (taxanes), the cell response to tubulin polymerization inhibitors remains less well characterized. Arylthioindoles (ATIs) are recently developed tubulin inhibitors. We previously identified ATI members that effectively inhibit tubulin polymerization in vitro and cancer cell growth in bulk cell viability assays. Here we characterise in depth the response of cancer cell lines to five selected ATIs. We find that all ATIs arrest mitotic progression, yet subsequently yield distinct cell fate profiles in time-lapse recording assays, indicating that molecules endowed with similar tubulin polymerization inhibitory activity in vitro can in fact display differential efficacy in living cells. Individual ATIs induce cytological phenotypes of increasing severity in terms of damage to the mitotic apparatus. That differentially triggers MCL-1 down-regulation and caspase-3 activation, and underlies the terminal fate of treated cells. Collectively, these results contribute to define the cell response to tubulin inhibitors and pinpoint potentially valuable molecules that can increase the molecular diversity of tubulin-targeting agents.
Collapse
|
45
|
Miura Y, Kaira K, Sakurai R, Imai H, Tomizawa Y, Sunaga N, Minato K, Hisada T, Oyama T, Yamada M. Prognostic effect of class III β-tubulin and Topoisomerase-II in patients with advanced thymic carcinoma who received combination chemotherapy, including taxanes or topoisomerase-II inhibitors. Oncol Lett 2017; 14:2369-2378. [PMID: 28789453 PMCID: PMC5530002 DOI: 10.3892/ol.2017.6419] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 01/31/2017] [Indexed: 01/03/2023] Open
Abstract
Class III β-tubulin (TUBB3) and Topoisomerase-II (topo-II) are considered to be the predictors of therapeutic efficacy and outcome in several types of human neoplasm. However, whether TUBB3 or topo-II may predict the response to combination chemotherapy and prognosis in patients with advanced thymic carcinoma (ATC) remains unclear. The aim of the present study was to investigate the prognostic significance of TUBB3 and topo-II expression levels in ATC. A total of 34 patients with ATC who received combination chemotherapy were enrolled in the present study. Immunohistochemical analysis was used to examine the expression of TUBB3, topo-II and Ki-67 in tumor specimens obtained by surgical resection or biopsy. TUBB3 and topo-II were highly expressed in 38 and 53% of the tumors, respectively. Progression-free survival (PFS) was significantly shorter in patients with high levels of TUBB3 compared with those with low levels of TUBB3 (P<0.01), whereas no significant difference in PFS between patients with high and low topo-II expression levels was observed (P=0.31). Patients with overexpression of TUBB3 or topo-II exhibited significantly shorter overall survival rates (OS) compared with those patients with low levels of expression of these proteins (TUBB3; P=0.01, topo-II; P=0.01). Multivariate analysis demonstrated that a high level of TUBB3 expression was an independent unfavorable prognostic factor for OS, and a high level of topo-II expression tended to correlate with poor prognosis without statistical significance. Additionally, a subset analysis demonstrated that the treatment with taxanes, but not topo-II inhibitors, tended to prolong OS in patients with TUBB3 overexpression and there was significant survival advantage of chemoradiotherapy over chemotherapy in patients with topo-II overexpression. It was revealed that an enhanced expression of TUBB3 or topo-II was clearly associated with clinical outcomes in patients with ATC who received combination chemotherapy, including taxanes or topo-II inhibitors, suggesting the prognostic significance of these markers.
Collapse
Affiliation(s)
- Yosuke Miura
- Division of Respiratory Medicine, Gunma Prefectural Cancer Center, Ota, Gunma 373-8550, Japan
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Kyoichi Kaira
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
- Department of Oncology Clinical Development, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Reiko Sakurai
- Division of Respiratory Medicine, National Hospital Organization Nishigunma Hospital, Shibukawa, Gunma 377-8511, Japan
| | - Hisao Imai
- Division of Respiratory Medicine, Gunma Prefectural Cancer Center, Ota, Gunma 373-8550, Japan
| | - Yoshio Tomizawa
- Division of Respiratory Medicine, National Hospital Organization Nishigunma Hospital, Shibukawa, Gunma 377-8511, Japan
| | - Noriaki Sunaga
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
- Oncology Center, Gunma University Hospital, Maebashi, Gunma 371-8511, Japan
| | - Koichi Minato
- Division of Respiratory Medicine, Gunma Prefectural Cancer Center, Ota, Gunma 373-8550, Japan
| | - Takeshi Hisada
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Tetsunari Oyama
- Department of Diagnostic Pathology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Masanobu Yamada
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
46
|
Liu X, Chang X, Liu R, Yu X, Chen L, Aihara K. Quantifying critical states of complex diseases using single-sample dynamic network biomarkers. PLoS Comput Biol 2017; 13:e1005633. [PMID: 28678795 PMCID: PMC5517040 DOI: 10.1371/journal.pcbi.1005633] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 07/19/2017] [Accepted: 06/19/2017] [Indexed: 02/04/2023] Open
Abstract
Dynamic network biomarkers (DNB) can identify the critical state or tipping point of a disease, thereby predicting rather than diagnosing the disease. However, it is difficult to apply the DNB theory to clinical practice because evaluating DNB at the critical state required the data of multiple samples on each individual, which are generally not available, and thus limit the applicability of DNB. In this study, we developed a novel method, i.e., single-sample DNB (sDNB), to detect early-warning signals or critical states of diseases in individual patients with only a single sample for each patient, thus opening a new way to predict diseases in a personalized way. In contrast to the information of differential expressions used in traditional biomarkers to “diagnose disease”, sDNB is based on the information of differential associations, thereby having the ability to “predict disease” or “diagnose near-future disease”. Applying this method to datasets for influenza virus infection and cancer metastasis led to accurate identification of the critical states or correct prediction of the immediate diseases based on individual samples. We successfully identified the critical states or tipping points just before the appearance of disease symptoms for influenza virus infection and the onset of distant metastasis for individual patients with cancer, thereby demonstrating the effectiveness and efficiency of our method for quantifying critical states at the single-sample level. The concept of dynamic network biomarkers (DNB) was proposed for detecting the critical state or tipping point of a complex disease (a pre-disease state immediately preceding the disease state), and has been applied to study the mechanism of cell fate decision and immune checkpoint blockade. But DNB cannot be used to identify the critical state or tipping point for a single patient because evaluating DNB for critical state required the data of multiple samples. The proposed method can identify the critical state of a complex disease for a single patient by implementing the concept of DNB. This method not only can be applied to detect the critical state or tipping point of a single sample, but also can be used to study the mechanism of complex disease at a single sample level. The ability of accurately and efficiently identifying the critical state for a single sample can benefit the development of personalized medicine.
Collapse
Affiliation(s)
- Xiaoping Liu
- Institute of Industrial Science, the University of Tokyo, Tokyo, Japan
- College of Statistics and Applied Mathematics, Anhui University of Finance and Economics, Bengbu, Anhui Province, China
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- School of Mathematics and Statistics, Shandong University at Weihai, Weihai, China
| | - Xiao Chang
- Institute of Industrial Science, the University of Tokyo, Tokyo, Japan
- College of Statistics and Applied Mathematics, Anhui University of Finance and Economics, Bengbu, Anhui Province, China
| | - Rui Liu
- School of Mathematics, South China University of Technology, Guangzhou, China
| | - Xiangtian Yu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Luonan Chen
- Institute of Industrial Science, the University of Tokyo, Tokyo, Japan
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- * E-mail: (LC); (KA)
| | - Kazuyuki Aihara
- Institute of Industrial Science, the University of Tokyo, Tokyo, Japan
- * E-mail: (LC); (KA)
| |
Collapse
|
47
|
Parker AL, Teo WS, McCarroll JA, Kavallaris M. An Emerging Role for Tubulin Isotypes in Modulating Cancer Biology and Chemotherapy Resistance. Int J Mol Sci 2017; 18:ijms18071434. [PMID: 28677634 PMCID: PMC5535925 DOI: 10.3390/ijms18071434] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 06/24/2017] [Accepted: 06/27/2017] [Indexed: 12/19/2022] Open
Abstract
Tubulin proteins, as components of the microtubule cytoskeleton perform critical cellular functions throughout all phases of the cell cycle. Altered tubulin isotype composition of microtubules is emerging as a feature of aggressive and treatment refractory cancers. Emerging evidence highlighting a role for tubulin isotypes in differentially influencing microtubule behaviour and broader functional networks within cells is illuminating a complex role for tubulin isotypes regulating cancer biology and chemotherapy resistance. This review focuses on the role of different tubulin isotypes in microtubule dynamics as well as in oncogenic changes that provide a survival or proliferative advantage to cancer cells within the tumour microenvironment and during metastatic processes. Consideration of the role of tubulin isotypes beyond their structural function will be essential to improving the current clinical use of tubulin-targeted chemotherapy agents and informing the development of more effective cancer therapies.
Collapse
Affiliation(s)
- Amelia L Parker
- Tumour Biology and Targeting, Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2031, Australia.
- Australian Centre for NanoMedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Wee Siang Teo
- Tumour Biology and Targeting, Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2031, Australia.
- Australian Centre for NanoMedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Joshua A McCarroll
- Tumour Biology and Targeting, Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2031, Australia.
- Australian Centre for NanoMedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Maria Kavallaris
- Tumour Biology and Targeting, Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2031, Australia.
- Australian Centre for NanoMedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
48
|
Cirillo L, Gotta M, Meraldi P. The Elephant in the Room: The Role of Microtubules in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1002:93-124. [DOI: 10.1007/978-3-319-57127-0_5] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
49
|
Smiyun G, Azarenko O, Miller H, Rifkind A, LaPointe NE, Wilson L, Jordan MA. βIII-tubulin enhances efficacy of cabazitaxel as compared with docetaxel. Cancer Chemother Pharmacol 2017; 80:151-164. [DOI: 10.1007/s00280-017-3345-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 05/24/2017] [Indexed: 11/28/2022]
|
50
|
Chemosensitization and inhibition of pancreatic cancer stem cell proliferation by overexpression of microRNA-205. Cancer Lett 2017; 402:1-8. [PMID: 28536008 DOI: 10.1016/j.canlet.2017.05.007] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/26/2017] [Accepted: 05/14/2017] [Indexed: 12/23/2022]
Abstract
Treatment of pancreatic cancer with gemcitabine (GEM) is limited due to its rapid plasma metabolism and development of chemoresistance. MicroRNA (miRNA) regulates cancer stem cell (CSC) maintenance and induces chemoresistance in cancer cells. In this study, we observed differential downregulation of miR-205 (miR-205-5p) in human pancreatic cancer tissues and cells. Compared to GEM-sensitive MIA PaCa-2 cells, miR-205 was highly downregulated in GEM-resistant MIA PaCa-2R cells. Lentivirus-mediated overexpression of miR-205 inhibits MIA PaCa-2R cell proliferation after GEM-treatment. Further investigation confirmed that miR-205 alone significantly reduces the proliferation of CSCs and tumor growth in mouse models. However, miR-205 in combination with GEM was more efficient in reducing the proliferation of CSCs and 3D spheroids. Moreover, miR-205 overexpressing MIA PaCa-2R cells induced orthotopic tumor growth was significantly inhibited after intravenous administration of GEM-conjugated methoxy poly(ethylene glycol)-block-poly(2-methyl-2-carboxyl-propylene carbonate)-graft-gemcitabine-graft-dodecanol (mPEG-b-PCC-g-GEM-g-DC) (mPEG-b-PCC-g-GEM-g-DC) polymeric micelles. Also, a reduction in CSCs, EMT and chemoresistance markers was observed in miR-205 overexpressing MIA PaCa-2R cells. Immunohistochemical analysis of orthotopic tumors showed a decrease in drug resistance protein caveolin-1 and cell proliferation marker Ki-67 in combination treatment. Overall, our findings suggest that miR-205 resensitizes GEM-resistant pancreatic cancer cells to GEM and acts as a tumor suppressor miRNA.
Collapse
|