1
|
Singh S, Sachan K, Verma S, Singh N, Singh PK. Cubosomes: An Emerging and Promising Drug Delivery System for Enhancing Cancer Therapy. Curr Pharm Biotechnol 2024; 25:757-771. [PMID: 37929730 DOI: 10.2174/0113892010257937231025065352] [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: 05/07/2023] [Revised: 08/26/2023] [Accepted: 09/01/2023] [Indexed: 11/07/2023]
Abstract
Cancer and other diseases can be treated with cubosomes, which are lyotropic nonlamellar liquid crystalline nanoparticles (LCNs). These cubosomes can potentially be a highly versatile carrier with theranostic efficacy, as they can be ingested, applied topically, or injected intravenously. Recent years have seen substantial progress in the synthesis, characterization, regulation of drug release patterns, and target selectivity of loaded anticancer bioactive compounds. However, its use in clinical settings has been slow and necessitates additional proof. Recent progress and roadblocks in using cubosomes as a nanotechnological intervention against various cancers are highlighted. In the last few decades, advances in biomedical nanotechnology have allowed for the development of "smart" drug delivery devices that can adapt to external stimuli. By improving therapeutic targeting efficacy and lowering the negative effects of payloads, these well-defined nanoplatforms can potentially promote patient compliance in response to specific stimuli. Liposomes and niosomes, two other well-known vesicular systems, share a lipid basis with cubosomes. Possible applications include a novel medication delivery system for hydrophilic, lipophilic, and amphiphilic drugs. We evaluate the literature on cubosomes, emphasizing their potential use in tumor-targeted drug delivery applications and critiquing existing explanations for cubosome self-assembly, composition, and production. As cubosome dispersion has bioadhesive and compatible features, numerous drug delivery applications, including oral, ocular, and transdermal, are also discussed in this review.
Collapse
Affiliation(s)
- Smita Singh
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology, Delhi NCR Campus, Modinagar, Ghaziabad, India
| | - Kapil Sachan
- KIET School of Pharmacy, KIET Group of Institutions, Ghaziabad, India
| | - Suryakant Verma
- School of Pharmacy, Bharat Institute of Technology, Meerut, India
| | - Nidhi Singh
- Sunder Deep Pharmacy College, Dasna, Ghaziabad, India
| | - Pranjal Kumar Singh
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology, Delhi NCR Campus, Modinagar, Ghaziabad, India
| |
Collapse
|
2
|
Meng Z, Chen J, Xu L, Xiao X, Zong L, Han Y, Jiang B. Study on Cytochrome P450 Metabolic Profile of Paclitaxel on Rats using QTOF-MS. Curr Drug Metab 2024; 25:330-339. [PMID: 39039675 DOI: 10.2174/0113892002308509240711100502] [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: 04/03/2024] [Revised: 06/05/2024] [Accepted: 06/10/2024] [Indexed: 07/24/2024]
Abstract
BACKGROUND Paclitaxel (PTX) is a key drug used for chemotherapy for various cancers. The hydroxylation metabolites of paclitaxel are different between humans and rats. Currently, there is little information available on the metabolic profiles of CYP450 enzymes in rats. OBJECTIVE This study evaluated the dynamic metabolic profiles of PTX and its metabolites in rats and in vitro. METHODS Ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UHPLC-Q-TOF-MS) and LC-MS/MS were applied to qualitative and quantitative analysis of PTX and its metabolites in rats, liver microsomes and recombinant enzyme CYP3A1/3A2. Ten specific inhibitors [NF (CYP1A1), FFL (CYP1A2), MOP (CYP2A6), OND (CYP2B6), QCT (CYP2C8), SFP (CYP2C9), NKT (CYP2C19), QND (CYP2D6), MPZ (CYP2E1) and KTZ (CYP3A4)] were used to identify the metabolic pathway in vitro. RESULTS Four main hydroxylated metabolites of PTX were identified. Among them, 3'-p-OH PTX and 2-OH PTX were monohydroxylated metabolites identified in rats and liver microsome samples, and 6α-2-di-OH PTX and 6α-5"-di-OH PTX were dihydroxylated metabolites identified in rats. CYP3A recombinant enzyme studies showed that the CYP3A1/3A2 in rat liver microsomes was mainly responsible for metabolizing PTX into 3'-p- OH-PTX and 2-OH-PTX. However, 6α-OH PTX was not detected in rat plasma and liver microsome samples. CONCLUSION The results indicated that the CYP3A1/3A2 enzyme, metabolizing PTX into 3'-p-OH-PTX and 2- OH-PTX, is responsible for the metabolic of PTX in rats. The CYP2C8 metabolite 6α-OH PTX in humans was not detected in rat plasma in this study, which might account for the interspecies metabolic differences between rats and humans. This study will provide evidence for drug-drug interaction research in rats.
Collapse
Affiliation(s)
- Zhaoyang Meng
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yi Shan Road, Shanghai, 200233, P. R. China
- College of Food Science and Technology, Shanghai Ocean University, 999 Hucheng Huan Road, Shanghai, 201306, P. R. China
| | - Junjun Chen
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yi Shan Road, Shanghai, 200233, P. R. China
| | - Lingyan Xu
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yi Shan Road, Shanghai, 200233, P. R. China
| | - Xiao Xiao
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yi Shan Road, Shanghai, 200233, P. R. China
| | - Ling Zong
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yi Shan Road, Shanghai, 200233, P. R. China
| | - Yonglong Han
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yi Shan Road, Shanghai, 200233, P. R. China
| | - Bo Jiang
- Department of Pharmacy, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 600 Yi Shan Road, Shanghai, 200233, P. R. China
| |
Collapse
|
3
|
Bhattacharya D, Mukhopadhyay M, Shivam K, Tripathy S, Patra R, Pramanik A. Recent developments in photodynamic therapy and its application against multidrug resistant cancers. Biomed Mater 2023; 18:062005. [PMID: 37827172 DOI: 10.1088/1748-605x/ad02d4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 10/12/2023] [Indexed: 10/14/2023]
Abstract
Recently, photodynamic therapy (PDT) has received a lot of attention for its potential use in cancer treatment. It enables the therapy of a multifocal disease with the least amount of tissue damage. The most widely used prodrug is 5-aminolevulinic acid, which undergoes heme pathway conversion to protoporphyrin IX, which acts as a photosensitizer (PS). Additionally, hematoporphyrin, bacteriochlorin, and phthalocyanine are also studied for their therapeutic potential in cancer. Unfortunately, not every patient who receives PDT experiences a full recovery. Resistance to different anticancer treatments is commonly observed. A few of the resistance mechanisms by which cancer cells escape therapeutics are genetic factors, drug-drug interactions, impaired DNA repair pathways, mutations related to inhibition of apoptosis, epigenetic pathways, etc. Recently, much research has been conducted to develop a new generation of PS based on nanomaterials that could be used to overcome cancer cells' multidrug resistance (MDR). Various metal-based, polymeric, lipidic nanoparticles (NPs), dendrimers, etc, have been utilized in the PDT application against cancer. This article discusses the detailed mechanism by which cancer cells evolve towards MDR as well as recent advances in PDT-based NPs for use against multidrug-resistant cancers.
Collapse
Affiliation(s)
- Debalina Bhattacharya
- Department of Microbiology, Maulana Azad College, Kolkata, West Bengal 700013, India
| | - Mainak Mukhopadhyay
- Department of Biotechnology, JIS University, Kolkata, West Bengal 700109, India
| | - Kumar Shivam
- Amity Institute of Click Chemistry Research & Studies, Amity University, Noida 201301, India
| | - Satyajit Tripathy
- Department of Pharmacology, University of Free State, Bloemfontein, Free State, 9301, South Africa
- Amity Institute of Allied Health Science, Amity University, Noida 201301, India
| | - Ranjan Patra
- Amity Institute of Click Chemistry Research & Studies, Amity University, Noida 201301, India
- Department of Chemistry, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Arindam Pramanik
- School of Medicine, University of Leeds, Leeds, LS9 7TF, United Kingdom
- Amity Institute of Biotechnology, Amity University, Noida 201301, India
| |
Collapse
|
4
|
Sofi FA, Tabassum N. Natural product inspired leads in the discovery of anticancer agents: an update. J Biomol Struct Dyn 2023; 41:8605-8628. [PMID: 36255181 DOI: 10.1080/07391102.2022.2134212] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 10/03/2022] [Indexed: 10/24/2022]
Abstract
Natural products have emerged as major leads for the discovery and development of new anti-cancer drugs. The plant-derived anti-cancer drugs account for approximately 60% and the quest for new anti-cancer agents is in progress. Anti-cancer leads have been isolated from plants, animals, marine organisms, and microorganisms from time immemorial. The process of semisynthetic modifications of the parent lead has led to the generation of new anti-cancer agents with improved therapeutic efficacy and minimal side effects. The various chemo-informatics tools, bioinformatics, high-throughput screening, and combinatorial synthesis are able to deliver the new natural product lead molecules. Plant-derived anticancer agents in either late preclinical development or early clinical trials include taxol, vincristine, vinblastine, topotecan, irinotecan, etoposide, paclitaxel, and docetaxel. Similarly, anti-cancer agents from microbial sources include dactinomycin, bleomycin, mitomycin C, and doxorubicin. In this review, we highlighted the importance of natural products leads in the discovery and development of novel anti-cancer agents. The semisynthetic modifications of the parent lead to the new anti-cancer agent are also presented. Further, the leads in the preclinical settings with the potential to become effective anticancer agents are also reviewed.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Firdoos Ahmad Sofi
- Department of Pharmaceutical Sciences, School of Applied Sciences and Technology, University of Kashmir, Srinagar, Jammu & Kashmir, India
| | - Nahida Tabassum
- Department of Pharmaceutical Sciences, School of Applied Sciences and Technology, University of Kashmir, Srinagar, Jammu & Kashmir, India
| |
Collapse
|
5
|
Choudhary N, Bawari S, Burcher JT, Sinha D, Tewari D, Bishayee A. Targeting Cell Signaling Pathways in Lung Cancer by Bioactive Phytocompounds. Cancers (Basel) 2023; 15:3980. [PMID: 37568796 PMCID: PMC10417502 DOI: 10.3390/cancers15153980] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/29/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Lung cancer is a heterogeneous group of malignancies with high incidence worldwide. It is the most frequently occurring cancer in men and the second most common in women. Due to its frequent diagnosis and variable response to treatment, lung cancer was reported as the top cause of cancer-related deaths worldwide in 2020. Many aberrant signaling cascades are implicated in the pathogenesis of lung cancer, including those involved in apoptosis (B cell lymphoma protein, Bcl-2-associated X protein, first apoptosis signal ligand), growth inhibition (tumor suppressor protein or gene and serine/threonine kinase 11), and growth promotion (epidermal growth factor receptor/proto-oncogenes/phosphatidylinositol-3 kinase). Accordingly, these pathways and their signaling molecules have become promising targets for chemopreventive and chemotherapeutic agents. Recent research provides compelling evidence for the use of plant-based compounds, known collectively as phytochemicals, as anticancer agents. This review discusses major contributing signaling pathways involved in the pathophysiology of lung cancer, as well as currently available treatments and prospective drug candidates. The anticancer potential of naturally occurring bioactive compounds in the context of lung cancer is also discussed, with critical analysis of their mechanistic actions presented by preclinical and clinical studies.
Collapse
Affiliation(s)
- Neeraj Choudhary
- Department of Pharmacognosy, GNA School of Pharmacy, GNA University, Phagwara 144 401, India
| | - Sweta Bawari
- Amity Institute of Pharmacy, Amity University, Noida 201 301, India
| | - Jack T. Burcher
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
| | - Dona Sinha
- Department of Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, Kolkata 700 026, India
| | - Devesh Tewari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi 110 017, India
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
| |
Collapse
|
6
|
PLGA-Lipid Hybrid Nanoparticles for Overcoming Paclitaxel Tolerance in Anoikis-Resistant Lung Cancer Cells. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238295. [PMID: 36500387 PMCID: PMC9737185 DOI: 10.3390/molecules27238295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/19/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022]
Abstract
Drug resistance and metastasis are two major obstacles to cancer chemotherapy. During metastasis, cancer cells can survive as floating cells in the blood or lymphatic circulatory system, due to the acquisition of resistance to anoikis-a programmed cell death activated by loss of extracellular matrix attachment. The anoikis-resistant lung cancer cells also develop drug resistance. In this study, paclitaxel-encapsulated PLGA-lipid hybrid nanoparticles (PLHNPs) were formulated by nanoprecipitation combined with self-assembly. The paclitaxel-PLHNPs had an average particle size of 103.0 ± 1.6 nm and a zeta potential value of -52.9 mV with the monodisperse distribution. Cytotoxicity of the nanoparticles was evaluated in A549 human lung cancer cells cultivated as floating cells under non-adherent conditions, compared with A549 attached cells. The floating cells exhibited anoikis resistance as shown by a lack of caspase-3 activation, in contrast to floating normal epithelial cells. Paclitaxel tolerance was evident in floating cells which had an IC50 value of 418.56 nM, compared to an IC50 value of 7.88 nM for attached cells. Paclitaxel-PLHNPs significantly reduced the IC50 values in both attached cells (IC50 value of 0.11 nM, 71.6-fold decrease) and floating cells (IC50 value of 1.13 nM, 370.4-fold decrease). This report demonstrated the potential of PLHNPs to improve the efficacy of the chemotherapeutic drug paclitaxel, for eradicating anoikis-resistant lung cancer cells during metastasis.
Collapse
|
7
|
S. M. S, Naveen NR, Rao GSNK, Gopan G, Chopra H, Park MN, Alshahrani MM, Jose J, Emran TB, Kim B. A spotlight on alkaloid nanoformulations for the treatment of lung cancer. Front Oncol 2022; 12:994155. [PMID: 36330493 PMCID: PMC9623325 DOI: 10.3389/fonc.2022.994155] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/12/2022] [Indexed: 07/30/2023] Open
Abstract
Numerous naturally available phytochemicals have potential anti-cancer activities due to their vast structural diversity. Alkaloids have been extensively used in cancer treatment, especially lung cancers, among the plant-based compounds. However, their utilization is limited by their poor solubility, low bioavailability, and inadequacies such as lack of specificity to cancer cells and indiscriminate distribution in the tissues. Incorporating the alkaloids into nanoformulations can overcome the said limitations paving the way for effective delivery of the alkaloids to the site of action in sufficient concentrations, which is crucial in tumor targeting. Our review attempts to assess whether alkaloid nanoformulation can be an effective tool in lung cancer therapy. The mechanism of action of each alkaloid having potential is explored in great detail in the review. In general, Alkaloids suppress oncogenesis by modulating several signaling pathways involved in multiplication, cell cycle, and metastasis, making them significant component of many clinical anti-cancerous agents. The review also explores the future prospects of alkaloid nanoformulation in lung cancer. So, in conclusion, alkaloid based nanoformulation will emerge as a potential gamechanger in treating lung cancer in the near future.
Collapse
Affiliation(s)
- Sindhoor S. M.
- Department of Pharmaceutics, P.A. College of Pharmacy, Mangalore, Karnataka, India
| | - N. Raghavendra Naveen
- Department of Pharmaceutics, Sri Adichunchanagiri College of Pharmacy, Adichunchanagiri University, B. G. Nagar, Karnataka, India
| | - GSN Koteswara Rao
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Gopika Gopan
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, Nitte (Deemed to be University), Mangalore, Karnataka, India
| | - Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Moon Nyeo Park
- Department of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Mohammed Merae Alshahrani
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Jobin Jose
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Sciences, Nitte (Deemed to be University), Mangalore, Karnataka, India
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Bonglee Kim
- Department of Korean Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
8
|
Kashio-Yokota Y, Sato S, Hirose T, Watanabe T, Endo A, Watanabe F, Endo M, Ohba K, Mori T, Takahashi K. Elevated (Pro)renin Receptor Expression by Anti-Cancer Drugs, Carboplatin and Paclitaxel, in Cultured Cancer Cells: Possible Involvement of Apoptosis and Autophagy. TOHOKU J EXP MED 2021; 255:91-104. [PMID: 34645770 DOI: 10.1620/tjem.255.91] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
(Pro)renin receptor [(P)RR] is a component of the renin-angiotensin system and plays an essential role in the activity of vacuolar H+-ATPase and autophagy. (P)RR is expressed in cancer cells. However, the relationship among (P)RR, apoptosis and autophagy in the treatment of anti-cancer drugs has not been clarified. The aim of this study was to clarify the effects of anti-cancer drugs with autophagy-promoting activity on (P)RR expression in cancer cells. MCF-7 breast cancer cells and A549 lung cancer cells were treated with carboplatin or paclitaxel, and the expression of (P)RR, apoptosis markers and autophagy markers were assessed by RT-qPCR, western blot analysis and immunocytochemistry. Expression levels of (P)RR mRNA and soluble (P)RR protein were increased by carboplatin or paclitaxel in a dose-dependent manner. Immunofluorescence staining of (P)RR was increased in both MCF-7 and A549 cells treated by carboplatin or paclitaxel. Apoptosis induction was shown by elevated BAX/BCL2 mRNA levels and increased active caspase3-positive cells. Moreover, autophagy induction was confirmed by increased levels of autophagy-associated mRNAs and LC3B-II proteins. (P)RR knockdown by (P)RR-specific siRNA suppressed the cell viability in MCF-7 cells and A549 cells under the treatment of carboplatin or paclitaxel, suggesting that (P)RR deficiency inhibits the proliferation of cancer cells in a pathway different from carboplatin or paclitaxel. The present study showed that the expression of (P)RR mRNA and soluble (P)RR was increased by anti-cancer drugs with autophagy-promoting activity. Upregulated (P)RR and autophagy may constitute a stress adaptation that protects cancer cells from apoptosis.
Collapse
Affiliation(s)
- Yurina Kashio-Yokota
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine
| | - Shigemitsu Sato
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine
| | - Takuo Hirose
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine.,Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University.,Division of Integrative Renal Replacement Therapy, Faculty of Medicine, Tohoku Medical and Pharmaceutical University
| | - Tomoki Watanabe
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine
| | - Akari Endo
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine.,Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University
| | - Fumihiko Watanabe
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine
| | - Moe Endo
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine
| | - Koji Ohba
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine
| | - Takefumi Mori
- Division of Nephrology and Endocrinology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University.,Division of Integrative Renal Replacement Therapy, Faculty of Medicine, Tohoku Medical and Pharmaceutical University
| | - Kazuhiro Takahashi
- Department of Endocrinology and Applied Medical Science, Tohoku University Graduate School of Medicine
| |
Collapse
|
9
|
Shin SB, Kim CH, Jang HR, Yim H. Combination of Inhibitors of USP7 and PLK1 has a Strong Synergism against Paclitaxel Resistance. Int J Mol Sci 2020; 21:E8629. [PMID: 33207738 PMCID: PMC7697005 DOI: 10.3390/ijms21228629] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 12/23/2022] Open
Abstract
USP7 is a promising target for the development of cancer treatments because of its high expression and the critical functions of its substrates in carcinogenesis of several different carcinomas. Here, we demonstrated the effectiveness of targeting USP7 in advanced malignant cells showing high levels of USP7, especially in taxane-resistant cancer. USP7 knockdown effectively induced cell death in several cancer cells of lung, prostate, and cervix. Depletion of USP7 induced multiple spindle pole formation in mitosis, and, consequently, resulted in mitotic catastrophe. When USP7 was blocked in the paclitaxel-resistant lung cancer NCI-H460TXR cells, which has resistance to mitotic catastrophe, NCI-H460TXR cells underwent apoptosis effectively. Furthermore, combination treatment with the mitotic kinase PLK1 inhibitor volasertib and the USP7 inhibitor P22077 showed a strong synergism through down-regulation of MDR1/ABCB1 in paclitaxel-resistant lung cancer. Therefore, we suggest USP7 is a promising target for cancer therapy, and combination therapy with inhibitors of PLK1 and USP7 may be valuable for treating paclitaxel-resistant cancers, because of their strong synergism.
Collapse
Affiliation(s)
| | | | | | - Hyungshin Yim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do 15588, Korea; (S.-B.S.); (C.-H.K.); (H.-R.J.)
| |
Collapse
|
10
|
Overcoming the diverse mechanisms of multidrug resistance in lung cancer cells by photodynamic therapy using pTHPP-loaded PLGA-lipid hybrid nanoparticles. Eur J Pharm Biopharm 2020; 149:218-228. [PMID: 32112893 DOI: 10.1016/j.ejpb.2020.02.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 02/20/2020] [Accepted: 02/24/2020] [Indexed: 01/10/2023]
Abstract
Multidrug resistance (MDR) and the spread of cancer cells (metastasis) are major causes leading to failure of cancer treatment. MDR can develop in two main ways, with differences in their mechanisms for drug resistance, first drug-selected MDR developing after chemotherapeutic treatment, and metastasis-associated MDR acquired by cellular adaptation to microenvironmental changes during metastasis. This study aims to use a nanoparticle-mediated photodynamic therapy (NPs/PDT) approach to overcome both types of MDR. A photosensitizer, 5,10,15,20-Tetrakis(4-hydroxy-phenyl)-21H,23H-porphine (pTHPP) was loaded into poly(D,L-lactide-co-glycolide) (PLGA)-lipid hybrid nanoparticles. The photocytotoxic effect of the nanoparticles was evaluated using two different MDR models established from one cell line, A549 human lung adenocarcinoma, including (1) A549RT-eto, a MDR cell line derived from A549 cells by drug-selection, and (2) detachment-induced MDR acquired by A549 cells when cultured as floating cells under non-adherent conditions, which mimic metastasizing cancer cells in the blood/lymphatic circulation. In the drug-selected MDR model, A549RT-eto cells displayed 17.4- and 1.8-fold resistance to Etoposide and Paclitaxel, respectively, compared to the A549 parental cells. In contrast to treatment with anticancer drugs, NPs/PDT with pTHPP-loaded nanoparticles resulted in equal photocytotoxic effect in A549RT-eto and parental cells. Intracellular pTHPP accumulation and light-induced superoxide anion generation were observed at similar levels in the two cell lines. The NPs/PDT killed A549RT-eto and parental cells through apoptosis as revealed by flow cytometry. In the metastasis-associated MDR model, A549 floating cells exhibited resistance to Etoposide (11.6-fold) and Paclitaxel (57.8-fold) compared to A549 attached cells, but the floating cells failed to show resistance against the photocytotoxic effect of the NPs/PDT. The MDR overcoming activity of NPs/PDT is mainly due to delivery ability of the PLGA-lipid hybrid nanoparticles. In conclusion, this work suggests that PLGA-lipid hybrid nanoparticles have potential in delivering photosensitizer or chemotherapeutic drug for treating both drug-selected and metastasis-associated MDR lung cancer cells.
Collapse
|
11
|
Sun H, Zhou X, Bao Y, Xiong G, Cui Y, Zhou H. Involvement of miR-4262 in paclitaxel resistance through the regulation of PTEN in non-small cell lung cancer. Open Biol 2019; 9:180227. [PMID: 31337279 PMCID: PMC6685930 DOI: 10.1098/rsob.180227] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 06/14/2019] [Indexed: 12/30/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is considered to be the primary cause of cancer-related mortalities worldwide. Paclitaxel (PTX), either as a monotherapy or in combination with other drugs, is an alternative therapy for advanced NSCLC. However, cancer cell resistance against PTX represents a major clinical problem. This study aimed to investigate the role and underlying mechanism of miR-4262 in PTX-resistant NSCLC. The levels of miR-4262 were analysed by quantitative reverse transcription polymerase chain reaction. A luciferase reporter assay and bioinformatics were used to explore the potential target gene of miR-4262. Regulation of miR-4262 and PTEN expressions in NSCLC was conducted by transfection. PTX-resistant A549 and H1299 cells were established by stepwise screening through increasing the PTX concentration in the cultures. In vivo, tumorigenesis experiments were used to explore the effects of miR-4262 and PTX. Cell proliferation, apoptosis and cell migration were detected using a CCK-8 assay, flow cytometry and Transwell migration assay, respectively. PI3 K/Akt pathway-related proteins were detected by western blot. miR-4262 expression was significantly upregulated in NSCLC tissues and cell lines, and miR-4262 targeted PTEN. In addition, miR-4262 induced PTX chemoresistance by promoting survival and migration in A549/PTX and H1299/PTX cells. Moreover, miR-4262 expression and PI3 K/Akt signalling pathway-related proteins were upregulated and PTEN was downregulated in A549/PTX and H1299/PTX. Our results indicate that miR-4262 enhances PTX resistance in NSCLC cells through targeting PTEN and activating the PI3 K/Akt signalling pathway. The inhibition of miR-4262 expression might be an improved treatment to overcome PTX resistance in NSCLC.
Collapse
Affiliation(s)
- Hongwen Sun
- Department of Thoracic Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, People's Republic of China
| | - Xiaoting Zhou
- Clinical Medicine 2015, Second Clinical Medical College of Fujian Medical University, Quanzhou, Fujian 362000, People's Republic of China
| | - Yanan Bao
- Department of Thoracic Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, People's Republic of China
| | - Guosheng Xiong
- Department of Thoracic Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, People's Republic of China
| | - Yue Cui
- Department of Thoracic Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, People's Republic of China
| | - Hua Zhou
- Department of Oncology Radiotherapy, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, People's Republic of China
| |
Collapse
|
12
|
Huang L, Hu C, Chao H, Wang R, Lu H, Li H, Chen H. miR-29c regulates resistance to paclitaxel in nasopharyngeal cancer by targeting ITGB1. Exp Cell Res 2019; 378:1-10. [DOI: 10.1016/j.yexcr.2019.02.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 02/14/2019] [Accepted: 02/15/2019] [Indexed: 01/20/2023]
|
13
|
Poly(ethylene glycol)-block-poly(d,l-lactic acid) micelles containing oligo(lactic acid) 8-paclitaxel prodrug: In Vivo conversion and antitumor efficacy. J Control Release 2019; 298:186-193. [PMID: 30790593 DOI: 10.1016/j.jconrel.2019.02.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 02/02/2019] [Accepted: 02/13/2019] [Indexed: 01/09/2023]
Abstract
Poly(ethylene glycol)-block-poly(d,l-lactic acid) (PEG-b-PLA) micelles affect drug solubilization, and a paclitaxel (PTX) loaded-PEG-b-PLA micelle (PTX-PM) is approved for cancer treatment due to injection safety and dose escalation (Genexol-PM®) compared to Taxol®. However, PTX-PM is unstable in blood, has rapid clearance, and causes dose-limiting toxicity. We have synthesized a prodrug for PTX (7-OH), using oligo(lactic acid) as a novel pro-moiety (o(LA)8-PTX) specifically for PEG-b-PLA micelles, gaining higher loading and slower release of o(LA)8-PTX over PTX. Notably, o(LA)8-PTX prodrug converts into PTX by a backbiting reaction in vitro, without requiring esterases. We hypothesize that o(LA)8-PTX-loaded PEG-b-PLA micelles (o(LA)8-PTX-PM) has a lower Cmax and higher plasma AUC than PTX-PM for improved therapeutic effectiveness. In Sprague-Dawley rats at 10 mg/kg, compared to o(LA)8-PTX-PM (10% w/w loading) and PTX-PM (10%), o(LA)8-PTX-PM (50% w/w loading) produces a 2- and 3-fold higher plasma AUC0-24 of PTX, lactic acid-PTX, and o(LA)2-PTX (o(LA)0-2-PTX), respectively. For o(LA)8-PTX-PM at 10 and 50% w/w loading, PTX and lactic acid-PTX are major bioactive metabolites, respectively. Fast prodrug conversion of o(LA)8-PTX in vivo versus in vitro (by backbiting) suggests that o(LA)8 is a good substrate for esterases. At 60 mg/kg (qwx3), o(LA)8-PTX-PM (50%) has higher antitumor activity than o(LA)8-PTX-PM (10%) and PTX-PM (10%) in a syngeneic 4T1-luc breast tumor model based on measurements of tumor volume, 4T1-luc breast tumor bioluminescence, and survival. Importantly, intravenous administration of o(LA)8-PTX-PM is well tolerated by BALB/c mice. In summary, oligo(lactic acid)8-PTX is more compatible than PTX with PEG-b-PLA micelles, more stable, and may expand the role of PEG-b-PLA micelles from "solubilizer" into "nanocarrier" for PTX as a next-generation taxane for cancer.
Collapse
|
14
|
Wang Z, Zheng Y, Fang Z. The clinical efficacy and safety of paclitaxel combined with avastin for NSCLC patients diagnosed with malignant pleural effusion. ACTA ACUST UNITED AC 2018; 64:230-233. [PMID: 29641777 DOI: 10.1590/1806-9282.64.03.230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 07/17/2017] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The current study aimed to investigate the clinical efficacy of paclitaxel combined with avastin for non-small cell lung cancer (NSCLC) patients diagnosed with malignant pleural effusion (MPE). METHOD Total of 33 patients diagnosed with NSCLC as well as malignant pleural effusion were included. All of them received paclitaxel (175 mg/m2) and avastin (5 mg/kg). Clinical efficacy was evaluated using the total response rate, overall survival, progression-free survival and changes in MPE volume. Adverse events and rates of toxicities were examined as well. RESULTS The total response rate reached 77% while the overall survival and the median progression-free survival were respectively 22.2 months and 8.4 months. Toxicities of grade 3-4 consisted of neutropenia in 57% of patients, anemia in 17% of them, febrile neutropenia in 11%, as well as anorexia in 7%. No treatment-correlated deaths were found. CONCLUSION Paclitaxel combined with avastin decreased MPE volume and increased survival rate of NSCLC patients via inhibiting vascular endothelial growth factor expression.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Respiratory Medicine, Ningbo NO, 7 Hospital, China
| | - Yisheng Zheng
- Department of Respiratory and Critical Care Medicine, Fuzong Clinical College of Fujian Medical University, Fuzhou General Hospital, China
| | - Zhenjian Fang
- Department of Respiratory Medicine, Fuding Hospital, China
| |
Collapse
|
15
|
Świerczewska M, Klejewski A, Brązert M, Kaźmierczak D, Iżycki D, Nowicki M, Zabel M, Januchowski R. New and Old Genes Associated with Primary and Established Responses to Paclitaxel Treatment in Ovarian Cancer Cell Lines. Molecules 2018; 23:molecules23040891. [PMID: 29649113 PMCID: PMC6017641 DOI: 10.3390/molecules23040891] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/04/2018] [Accepted: 04/10/2018] [Indexed: 01/10/2023] Open
Abstract
Development of drug resistance is the main reason for low chemotherapy effectiveness in treating ovarian cancer. Paclitaxel (PAC) is a chemotherapeutic drug used in the treatment of this cancer. We analysed the development of PAC resistance in two ovarian cancer cell lines. Exposure of drug-sensitive cell lines (A2780 and W1) to PAC was used to determine the primary response. An established response was determined in PAC-resistant sublines of the A2780 and W1 cell lines. qRT-PCR was performed to measure the expression levels of specific genes. We observed decreased expression of the PCDH9, NSBP1, MCTP1 and SEMA3A genes in the PAC-resistant cell lines. Short-term exposure to PAC led to increased expression of the MDR1 and BCRP genes in the A2780 and W1 cell lines. In the A2780 cell line, we also observed increased expression of the C4orf18 gene and decreased expression of the PCDH9 and SEMA3A genes after PAC treatment. In the W1 cell line, short-term treatment with PAC upregulated the expression of the ALDH1A1 gene, a marker of Cancer stem cells (CSCs). Our results suggest that downregulation of the PCDH9, NSBP1, MCTP1 and SEMA3A genes and upregulation of the MDR1, BCRP, C4orf18 and ALDH1A1 genes may be related to PAC resistance.
Collapse
Affiliation(s)
- Monika Świerczewska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznań, Poland.
| | - Andrzej Klejewski
- Department of Nursing, Poznan University of Medical Sciences, Smoluchowskiego 11 St., 60-179 Poznan, Poland.
- Department of Obstetrics and Women's Diseases, Poznan University of Medical Sciences, Smoluchowskiego 11 St., 60-179 Poznan, Poland.
| | - Maciej Brązert
- Division of Infertility and Reproductive Endocrinology, Department of Gynecology, Obstetrics and Gynecological Oncology, Poznan University of Medical Sciences, Polna 33 St., 60-535 Poznań, Poland.
| | - Dominika Kaźmierczak
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznań, Poland.
| | - Dariusz Iżycki
- Department of Cancer Immunology, Poznan University of Medical Sciences, Garbary 15 St., 61-866 Poznań, Poland.
| | - Michał Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznań, Poland.
| | - Maciej Zabel
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznań, Poland.
| | - Radosław Januchowski
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznań, Poland.
| |
Collapse
|
16
|
Huang CY, Ju DT, Chang CF, Muralidhar Reddy P, Velmurugan BK. A review on the effects of current chemotherapy drugs and natural agents in treating non-small cell lung cancer. Biomedicine (Taipei) 2017; 7:23. [PMID: 29130448 PMCID: PMC5682982 DOI: 10.1051/bmdcn/2017070423] [Citation(s) in RCA: 270] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 09/20/2017] [Indexed: 12/20/2022] Open
Abstract
Lung cancer is the leading cause of cancer deaths worldwide, and this makes it an attractive disease to review and possibly improve therapeutic treatment options. Surgery, radiation, chemotherapy, targeted treatments, and immunotherapy separate or in combination are commonly used to treat lung cancer. However, these treatment types may cause different side effects, and chemotherapy-based regimens appear to have reached a therapeutic plateau. Hence, effective, better-tolerated treatments are needed to address and hopefully overcome this conundrum. Recent advances have enabled biologists to better investigate the potential use of natural compounds for the treatment or control of various cancerous diseases. For the past 30 years, natural compounds have been the pillar of chemotherapy. However, only a few compounds have been tested in cancerous patients and only partial evidence is available regarding their clinical effectiveness. Herein, we review the research on using current chemotherapy drugs and natural compounds (Wortmannin and Roscovitine, Cordyceps militaris, Resveratrol, OSU03013, Myricetin, Berberine, Antroquinonol) and the beneficial effects they have on various types of cancers including non-small cell lung cancer. Based on this literature review, we propose the use of these compounds along with chemotherapy drugs in patients with advanced and/or refractory solid tumours.
Collapse
Affiliation(s)
- Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 404, Taiwan - Graduate Institute of Chinese Medical Science, China Medical University, Taichung 404, Taiwan - Department of Biological Science and Technology, Asia University, Taichung 413, Taiwan
| | - Da-Tong Ju
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Chih-Fen Chang
- Department of Internal Medicine, Division of Cardiology, Armed Forces Taichung General Hospital, Taichung 406, Taiwan
| | - P Muralidhar Reddy
- Department of Chemistry, Nizam College, Osmania University, Hyderabad-500001, India
| | - Bharath Kumar Velmurugan
- Faculty of Applied Sciences, Ton Duc Thang University, Tan Phong Ward, District 7, 700000 Ho Chi Minh City, Vietnam
| |
Collapse
|
17
|
Wang S, Zhang S, Zhao Z, Zhang C, Yang X, Wang Y. Connexin 43 enhances paclitaxel cytotoxicity in colorectal cancer cell lines. Exp Ther Med 2017; 14:1212-1218. [PMID: 28810580 PMCID: PMC5526126 DOI: 10.3892/etm.2017.4589] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 04/07/2017] [Indexed: 01/08/2023] Open
Abstract
Colorectal cancer has a relatively low sensitivity to paclitaxel. The purpose of this study was to investigate the role of connexin 43 (Cx43), which is a structural component of gap junctional communication (GJC), in paclitaxel cytotoxicity in colorectal cancer cells. Three colorectal cancer cell lines (HCT106, HCT116 and LoVo) were transfected with Cx43 and used to examine paclitaxel cytotoxicity. A western blot assay was used to confirm Cx43 expression in transfected cell lines as well as the expression of several proteins that are associated with paclitaxel cytotoxicity. A parachute dye-coupling assay was used to measure GJC function. An MTT assay was used to analyze the viability of paclitaxel-treated cells. Cx43 expression level and GJC function were significantly upregulated by the transfection (P<0.05). The viability of transfected cells was significantly inhibited compared with that of untransfected cells when treated with paclitaxel (20 or 80 nM) at high culture density but not at low culture density (P<0.05). Cx43 transfection significantly increased the mitotic arrest, tubulin polymerization and apoptosis effects of paclitaxel (P<0.05). It was also found that paclitaxel had an inhibitory effect on GJC function after 12 h of treatment in LoVo cells (P<0.05). These results indicate that Cx43 may serve as a target of paclitaxel chemotherapy for colorectal cancer.
Collapse
Affiliation(s)
- Siqi Wang
- Laboratory of Biomedicine and Nanophotonics, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| | - Zhenying Zhao
- Department of Pharmacy, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| | - Chunze Zhang
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| | - Xiaoyun Yang
- Department of Pathology, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| | - Yijia Wang
- Laboratory of Biomedicine and Nanophotonics, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| |
Collapse
|
18
|
miR-30a-5p enhances paclitaxel sensitivity in non-small cell lung cancer through targeting BCL-2 expression. J Mol Med (Berl) 2017; 95:861-871. [PMID: 28487996 DOI: 10.1007/s00109-017-1539-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/20/2017] [Accepted: 04/27/2017] [Indexed: 12/13/2022]
Abstract
Lung cancer remains the leading cause of cancer-related death worldwide. Paclitaxel, either as monotherapy or combined with other agents, is the standard treatment for advanced non-small cell lung cancer (NSCLC), the most common type of lung cancer. However, both de novo and acquired resistance against paclitaxel frequently occurs and represents a huge clinical problem. The underlying mechanisms remain poorly characterized. Here, by comparing microRNA (miRNA) expression levels using miRNA arrays, we observed differential expression of miR-30a-5p in two independent lung cancer cell pairs (paclitaxel-resistant vs paclitaxel-sensitive A549 cell lines). Overexpression of miR-30a-5p sensitizes NSCLC cells to paclitaxel both in vitro and in vivo. In addition, miR-30a-5p increases paclitaxel sensitivity by promoting chemotherapy-induced apoptosis via downregulating BCL-2, a key apoptosis regulator. High miR-30a-5p expression is positively correlated with enhanced responsiveness to paclitaxel and predicts a more favorable clinical outcome in NSCLC patients. Moreover, miR-30a-5p expression is negatively correlated with BCL-2 expression in NSCLC tissues. These data indicate that miR-30a-5p may be useful to treat paclitaxel-resistant lung cancer and may also provide a biomarker to predict paclitaxel responsiveness in lung cancer. KEY MESSAGES BCL-2 is a novel direct target of miR-30a-5p. miR-30a-5p enhances NSCLC paclitaxel sensitivity in vitro and in vivo. miR-30a-5p sensitizes NSCLC cells to paclitaxel by inducing apoptosis through BCL-2 inhibition. miR-30a-5p negatively correlates with BCL-2 and predicts a favorable clinical outcome in NSCLC patients.
Collapse
|
19
|
Darby LM, Meng H, Fehrenbacher JC. Paclitaxel inhibits the activity and membrane localization of PKCα and PKCβI/II to elicit a decrease in stimulated calcitonin gene-related peptide release from cultured sensory neurons. Mol Cell Neurosci 2017; 82:105-117. [PMID: 28404507 DOI: 10.1016/j.mcn.2017.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/08/2017] [Accepted: 04/08/2017] [Indexed: 11/27/2022] Open
Abstract
Peripheral neuropathy is a dose-limiting and debilitating side effect of the chemotherapeutic drug, paclitaxel. Consequently, elucidating the mechanisms by which this drug alters sensory neuronal function is essential for the development of successful therapeutics for peripheral neuropathy. We previously demonstrated that chronic treatment with paclitaxel (3-5days) reduces neuropeptide release stimulated by agonists of TRPV1. Because the activity of TRPV1 channels is modulated by conventional and novel PKC isozymes (c/nPKC), we investigated whether c/nPKC mediate the loss of neuropeptide release following chronic treatment with paclitaxel (300nM; 3 and 5days). Release of the neuropeptide, calcitonin gene-related peptide (CGRP), was measured as an index of neuronal sensitivity. Following paclitaxel treatment, cultured dorsal root ganglia sensory neurons were stimulated with a c/nPKC activator, phorbol 12,13-dibutyrate (PDBu), or a TRPV1 agonist, capsaicin, in the absence and presence of selective inhibitors of conventional PKCα and PKCβI/II isozymes (cPKC). Paclitaxel (300nM; 3days and 5days) attenuated both PDBu- and capsaicin-stimulated release in a cPKC-dependent manner. Under basal conditions, there were no changes in the protein expression, phosphorylation or membrane localization of PKC α, βI or βII, however, paclitaxel decreased cPKC activity as indicated by a reduction in the phosphorylation of cPKC substrates. Under stimulatory conditions, paclitaxel attenuated the membrane translocation of phosphorylated PKC α, βI and βII, providing a rationale for the attenuation in PDBu- and capsaicin-stimulated release. Our findings suggest that a decrease in cPKC activity and membrane localization are responsible for the reduction in stimulated peptide release following chronic treatment with paclitaxel in sensory neurons.
Collapse
Affiliation(s)
- Lisa M Darby
- Indiana University School of Medicine, Department of Pharmacology and Toxicology, USA.
| | - Hongdi Meng
- Indiana University School of Medicine, Department of Pharmacology and Toxicology, USA
| | - Jill C Fehrenbacher
- Indiana University School of Medicine, Department of Pharmacology and Toxicology, USA; Indiana University School of Medicine, Stark Neuroscience Research Institute, USA.
| |
Collapse
|
20
|
Liao W, Zhao S, Zhang M, Dong K, Chen Y, Fu C, Yu L. Transcriptome Assembly and Systematic Identification of Novel Cytochrome P450s in Taxus chinensis. FRONTIERS IN PLANT SCIENCE 2017; 8:1468. [PMID: 28878800 PMCID: PMC5572210 DOI: 10.3389/fpls.2017.01468] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 08/07/2017] [Indexed: 05/06/2023]
Abstract
Taxus spp. is a highly valuable medicinal plant with multiple pharmacological effects on various cancers. Cytochrome P450s (CYP450s) play important roles in the biosynthesis of active compounds in Taxus spp., such as the famous diterpenoid, Taxol. However, some specific CYP450 enzymes involved in the biosynthesis of Taxol remain unknown, and the systematic identification of CYP450s in Taxus has not been reported. In this study, 118 full-length and 175 partial CYP450 genes were identified in Taxus chinensis transcriptomes. The 118 full-length genes were divided into 8 clans and 29 families. The CYP71 clan included all A-type genes (52) belonging to 11 families. The other seven clans possessed 18 families containing 66 non-A-type genes. Two new gymnosperm-specific families were discovered, and were named CYP864 and CYP947 respectively. Protein sequence alignments revealed that all of the T. chinensis CYP450s hold distinct conserved domains. The expression patterns of all 118 CYP450 genes during the long-time subculture and MeJA elicitation were analyzed. Additionally, the expression levels of 15 novel CYP725 genes in different Taxus species were explored. Considering all the evidence, 6 CYP725s were identified to be candidates for Taxol biosynthesis. The cis-regulatory elements involved in the transcriptional regulation were also identified in the promoter regions of CYP725s. This study presents a comprehensive overview of the CYP450 gene family in T. chinensis and can provide important insights into the functional gene studies of Taxol biosynthesis.
Collapse
Affiliation(s)
- Weifang Liao
- Department of Biotechnology, Institute of Resource Biology and Biotechnology, College of Life Science and Technology, Huazhong University of Science and TechnologyWuhan, China
| | - Shengying Zhao
- Department of Biotechnology, Institute of Resource Biology and Biotechnology, College of Life Science and Technology, Huazhong University of Science and TechnologyWuhan, China
| | - Meng Zhang
- Department of Biotechnology, Institute of Resource Biology and Biotechnology, College of Life Science and Technology, Huazhong University of Science and TechnologyWuhan, China
| | - Kaige Dong
- Department of Biotechnology, Institute of Resource Biology and Biotechnology, College of Life Science and Technology, Huazhong University of Science and TechnologyWuhan, China
| | - Ying Chen
- Department of Biotechnology, Institute of Resource Biology and Biotechnology, College of Life Science and Technology, Huazhong University of Science and TechnologyWuhan, China
| | - Chunhua Fu
- Department of Biotechnology, Institute of Resource Biology and Biotechnology, College of Life Science and Technology, Huazhong University of Science and TechnologyWuhan, China
- Key Laboratory of Molecular Biophysics Ministry of Education, College of Life Science and Technology, Huazhong University of Science and TechnologyWuhan, China
- *Correspondence: Chunhua Fu
| | - Longjiang Yu
- Department of Biotechnology, Institute of Resource Biology and Biotechnology, College of Life Science and Technology, Huazhong University of Science and TechnologyWuhan, China
- Key Laboratory of Molecular Biophysics Ministry of Education, College of Life Science and Technology, Huazhong University of Science and TechnologyWuhan, China
- Longjiang Yu
| |
Collapse
|
21
|
|
22
|
Zhao Y, Yang X. Regulation of sensitivity of tumor cells to antitubulin drugs by Cdk1-TAZ signalling. Oncotarget 2016; 6:21906-17. [PMID: 26183396 PMCID: PMC4673135 DOI: 10.18632/oncotarget.4259] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 05/22/2015] [Indexed: 12/22/2022] Open
Abstract
Antitubulin drugs are commonly used for the treatment of numerous cancers. However, either the intrinsic or acquired resistances of patients to these drugs result in the failure of the treatment and high mortality of cancers. Therefore, identifying genes or signalling pathways involved in antitubulin drug resistances is critical for future successful treatment of cancers. TAZ (Transcriptional coactivator with PDZ-binding motif), which is a core component of the Hippo pathway, is overexpressed in various cancers. We have recently shown that high levels of TAZ in cancer cells result in Taxol resistance through up-regulation of downstream targets Cyr61 and CTGF. However, how TAZ is regulated in response to Taxol is largely unknown. In this study, we found that Cdk1 (Cyclin-dependent kinase 1) directly phosphorylated TAZ on six novel sites independent of the Hippo pathway, which further resulted in TAZ degradation through proteasome system. Phosphorylation-mimicking TAZ mutant was unstable, and therefore abolished TAZ-induced antitubulin drug resistances. This study provides first evidence that Cdk1 is a novel kinase phosphorylating and regulating TAZ stability and suggests that Cdk1-TAZ signalling is a critical regulator of antitubulin drug response in cancer cells and may be a potential target for the treatment of antitubulin-drug resistant cancer patients.
Collapse
Affiliation(s)
- Yulei Zhao
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L3N6, Canada
| | - Xiaolong Yang
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L3N6, Canada
| |
Collapse
|
23
|
Recent advances in design, synthesis and bioactivity of paclitaxel-mimics. Fitoterapia 2016; 110:26-37. [DOI: 10.1016/j.fitote.2016.02.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 11/18/2022]
|
24
|
Abstract
Non-small cell lung cancer has seen an unprecedented augmentation of therapeutic options over the last couple of years. Improved understanding of molecular drivers and the role of the immune system in cancer therapy have brought new drugs to the armamentarium. Despite these advances, cytotoxic chemotherapy remains a substantial part of therapy for most patients in locally advanced and metastatic stage. Initially thought to be a chemotherapy-resistant entity, meta-analyses in the mid-1990s demonstrated modest efficacy of platinum-based therapy. Further combination trials demonstrated enhanced efficacy for several regimen in first and second lines, including the introduction of antimetabolites, taxanes, and anti-angiogenic agents. Maintenance chemotherapy has been another novel, successful approach for management of metastatic disease. Herein, we summarize the current concepts of chemotherapy, its applicability to the different histologies, and novel concepts of therapy.
Collapse
Affiliation(s)
- Martin F Dietrich
- Harold C. Simmons Cancer Center, Division of Hematology-Oncology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, 8852, Dallas, TX, 75390-8852, USA.
| | - David E Gerber
- Harold C. Simmons Cancer Center, Division of Hematology-Oncology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, 8852, Dallas, TX, 75390-8852, USA.
| |
Collapse
|
25
|
PRONOUNCE: randomized, open-label, phase III study of first-line pemetrexed + carboplatin followed by maintenance pemetrexed versus paclitaxel + carboplatin + bevacizumab followed by maintenance bevacizumab in patients ith advanced nonsquamous non-small-cell lung cancer. J Thorac Oncol 2015; 10:134-42. [PMID: 25371077 PMCID: PMC4276572 DOI: 10.1097/jto.0000000000000366] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION PRONOUNCE compared the efficacy and safety of pemetrexed+carboplatin followed by pemetrexed (Pem+Cb) with paclitaxel+carboplatin+bevacizumab followed by bevacizumab (Pac+Cb+Bev) in patients with advanced nonsquamous non-small-cell lung cancer (NSCLC). METHODS Patients ≥18 years of age with stage IV nonsquamous NSCLC (American Joint Committee on Cancer v7.0), and Eastern Cooperative Oncology Group performance status 0/1 were randomized (1:1) to four cycles of induction Pem+Cb (pemetrexed, 500 mg/m, carboplatin, area under the curve = 6) followed by Pem maintenance or Pac+Cb+Bev (paclitaxel, 200 mg/m, carboplatin, area under the curve = 6, and bevacizumab, 15 mg/kg) followed by Bev maintenance in the absence of progressive disease or discontinuation. The primary objective was progression-free survival (PFS) without grade 4 toxicity (G4PFS). Secondary end points were PFS, overall survival (OS), overall response rate (ORR), disease control rate (DCR), and safety. Resource utilization was also assessed. RESULTS Baseline characteristics of the patients randomized to Pem+Cb (N = 182) and Pac+Cb+Bev (N = 179) were well balanced between the arms. Median (months) G4PFS was 3.91 for Pem+Cb and 2.86 for Pac+Cb+Bev (hazard ratio = 0.85, 90% confidence interval, 0.7-1.04; p = 0.176); PFS, OS, ORR, or DCR did not differ significantly between the arms. Significantly more drug-related grade 3/4 anemia (18.7% versus 5.4%) and thrombocytopenia (24.0% versus 9.6%) were reported for Pem+Cb. Significantly more grade 3/4 neutropenia (48.8% versus 24.6%), grade 1/2 alopecia (28.3% versus 8.2%), and grade 1/2 sensory neuropathy were reported for Pac+Cb+Bev. Number of hospitalizations and overall length of stay did not differ significantly between the arms. CONCLUSIONS Pem+Cb did not produce significantly better G4PFS compared with Pac+Cb+Bev. Pem+Cb was not superior in PFS, OS, ORR, or DCR compared with Pac+Cb+Bev. Both regimens were well tolerated, although, toxicity profiles differed.
Collapse
|
26
|
Wu S, Liu X, Zhou X, Liang XM, Gao D, Liu H, Zhao G, Zhang Q, Wu X. Quantification of cell viability and rapid screening anti-cancer drug utilizing nanomechanical fluctuation. Biosens Bioelectron 2015; 77:164-73. [PMID: 26406457 DOI: 10.1016/j.bios.2015.09.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Revised: 08/31/2015] [Accepted: 09/10/2015] [Indexed: 11/27/2022]
Abstract
Cancer is a serious threat to human health. Although numerous anti-cancer drugs are available clinically, many have shown toxic side effects due to poor tumor-selectivity, and reduced effectiveness due to cancers rapid development of resistance to treatment. The development of new highly efficient and practical methods to quantify cell viability and its change under drug treatment is thus of significant importance in both understanding of anti-cancer mechanism and anti-cancer drug screening. Here, we present an approach of utilizing a nanomechanical fluctuation based highly sensitive microcantilever sensor, which is capable of characterizing the viability of cells and quantitatively screening (within tens of minutes) their responses to a drug with the obvious advantages of a rapid, label-free, quantitative, noninvasive, real-time and in-situ assay. The microcantilever sensor operated in fluctuation mode was used in evaluating the paclitaxel effectiveness on breast cancer cell line MCF-7. This study demonstrated that the nanomechanical fluctuations of the microcantilever sensor are sensitive enough to detect the dynamic variation in cellular force which is provided by the cytoskeleton, using cell metabolism as its energy source, and the dynamic instability of microtubules plays an important role in the generation of the force. We propose that cell viability consists of two parts: biological viability and mechanical viability. Our experimental results suggest that paclitaxel has little effect on biological viability, but has a significant effect on mechanical viability. This new method provides a new concept and strategy for the evaluation of cell viability and the screening of anti-cancer drugs.
Collapse
Affiliation(s)
- Shangquan Wu
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei 230027, China; Center for Biomedical Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Xiaoli Liu
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei 230026, China; Center for Biomedical Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Xiarong Zhou
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei 230027, China
| | - Xin M Liang
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Dayong Gao
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98195, USA
| | - Hong Liu
- Department of Chemical Physics, University of Science and Technology of China, Hefei 230026, China.
| | - Gang Zhao
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei 230026, China; Center for Biomedical Engineering, University of Science and Technology of China, Hefei 230026, China.
| | - Qingchuan Zhang
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei 230027, China; Center for Biomedical Engineering, University of Science and Technology of China, Hefei 230026, China.
| | - Xiaoping Wu
- CAS Key Laboratory of Mechanical Behavior and Design of Material, Department of Modern Mechanics, University of Science and Technology of China, Hefei 230027, China
| |
Collapse
|
27
|
Atjanasuppat K, Lirdprapamongkol K, Jantaree P, Svasti J. Non-adherent culture induces paclitaxel resistance in H460 lung cancer cells via ERK-mediated up-regulation of βIVa-tubulin. Biochem Biophys Res Commun 2015; 466:493-8. [PMID: 26375501 DOI: 10.1016/j.bbrc.2015.09.057] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 09/10/2015] [Indexed: 01/27/2023]
Abstract
Circulating tumor cells (CTCs) are metastasizing epithelial cancer cells that adapt to survive when floating in bloodstream during metastasis. This condition can be mimicked in vitro by using non-adherent cell culture. The chemosensitivity of CTCs appears to correlate with the response of metastatic cancer patients to therapy, but chemoresistance is also frequently observed in advanced stage cancer patients, who have never previously received chemotherapy. We hypothesize that adaptation of epithelial cancer cells to become floating CTCs could lead to development of chemoresistance. Here, we explore whether chemoresistance is induced in epithelial cancer cells when cultured under non-adherent conditions. Increased paclitaxel-specific resistance was observed in floating cells compared to attached cells in H460, MCF-7, and HepG2 human cancer cell lines, by 15.6-, 3.9-, and 2.6-fold increases in IC50 values, respectively. qRT-PCR analysis showed that a paclitaxel-resistant β-tubulin isotype, βIVa-tubulin, was the most up-regulated gene compared with other β-tubulin isotypes in H460 floating cells, concomitant with elevated ERK activation. ERK inhibitor treatment could attenuate the up-regulation of βIVa-tubulin, and decreased the paclitaxel resistance of H460 floating cells, even though other β-tubulin isotypes were up-regulated when the ERK activation was blocked. In conclusion, we show induction of paclitaxel resistance in epithelial cancer cells, when floating in non-adherent culture, and this might occur with CTCs of cancer patients.
Collapse
Affiliation(s)
- Korakot Atjanasuppat
- Applied Biological Sciences, Chulabhorn Graduate Institute, Bangkok 10210, Thailand
| | - Kriengsak Lirdprapamongkol
- Applied Biological Sciences, Chulabhorn Graduate Institute, Bangkok 10210, Thailand; Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok 10210, Thailand; Center of Excellence on Environmental Health and Toxicology (EHT), CHE, Ministry of Education, Bangkok 10400, Thailand.
| | - Phatcharida Jantaree
- Applied Biological Sciences, Chulabhorn Graduate Institute, Bangkok 10210, Thailand
| | - Jisnuson Svasti
- Applied Biological Sciences, Chulabhorn Graduate Institute, Bangkok 10210, Thailand; Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok 10210, Thailand
| |
Collapse
|
28
|
Mumenthaler SM, Foo J, Choi NC, Heise N, Leder K, Agus DB, Pao W, Michor F, Mallick P. The Impact of Microenvironmental Heterogeneity on the Evolution of Drug Resistance in Cancer Cells. Cancer Inform 2015; 14:19-31. [PMID: 26244007 PMCID: PMC4504404 DOI: 10.4137/cin.s19338] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 05/03/2015] [Accepted: 05/12/2015] [Indexed: 12/15/2022] Open
Abstract
Therapeutic resistance arises as a result of evolutionary processes driven by dynamic feedback between a heterogeneous cell population and environmental selective pressures. Previous studies have suggested that mutations conferring resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKI) in non-small-cell lung cancer (NSCLC) cells lower the fitness of resistant cells relative to drug-sensitive cells in a drug-free environment. Here, we hypothesize that the local tumor microenvironment could influence the magnitude and directionality of the selective effect, both in the presence and absence of a drug. Using a combined experimental and computational approach, we developed a mathematical model of preexisting drug resistance describing multiple cellular compartments, each representing a specific tumor environmental niche. This model was parameterized using a novel experimental dataset derived from the HCC827 erlotinib-sensitive and -resistant NSCLC cell lines. We found that, in contrast to in the drug-free environment, resistant cells may hold a fitness advantage compared to parental cells in microenvironments deficient in oxygen and nutrients. We then utilized the model to predict the impact of drug and nutrient gradients on tumor composition and recurrence times, demonstrating that these endpoints are strongly dependent on the microenvironment. Our interdisciplinary approach provides a model system to quantitatively investigate the impact of microenvironmental effects on the evolutionary dynamics of tumor cells.
Collapse
Affiliation(s)
- Shannon M Mumenthaler
- Center for Applied Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jasmine Foo
- School of Mathematics, University of Minnesota, Minneapolis, MN, USA
| | - Nathan C Choi
- Center for Applied Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Nicholas Heise
- School of Mathematics, University of Minnesota, Minneapolis, MN, USA
| | - Kevin Leder
- Department of Industrial and Systems Engineering, University of Minnesota, Minneapolis, MN, USA
| | - David B Agus
- Center for Applied Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - William Pao
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Franziska Michor
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Parag Mallick
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Palo Alto, CA, USA
| |
Collapse
|
29
|
Dasari S, Tchounwou PB. Cisplatin in cancer therapy: molecular mechanisms of action. Eur J Pharmacol 2014; 740:364-78. [PMID: 25058905 PMCID: PMC4146684 DOI: 10.1016/j.ejphar.2014.07.025] [Citation(s) in RCA: 3668] [Impact Index Per Article: 333.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 07/13/2014] [Accepted: 07/14/2014] [Indexed: 02/06/2023]
Abstract
Cisplatin, cisplatinum, or cis-diamminedichloroplatinum (II), is a well-known chemotherapeutic drug. It has been used for treatment of numerous human cancers including bladder, head and neck, lung, ovarian, and testicular cancers. It is effective against various types of cancers, including carcinomas, germ cell tumors, lymphomas, and sarcomas. Its mode of action has been linked to its ability to crosslink with the purine bases on the DNA; interfering with DNA repair mechanisms, causing DNA damage, and subsequently inducing apoptosis in cancer cells. However, because of drug resistance and numerous undesirable side effects such as severe kidney problems, allergic reactions, decrease immunity to infections, gastrointestinal disorders, hemorrhage, and hearing loss especially in younger patients, other platinum-containing anti-cancer drugs such as carboplatin, oxaliplatin and others, have also been used. Furthermore, combination therapies of cisplatin with other drugs have been highly considered to overcome drug-resistance and reduce toxicity. This comprehensive review highlights the physicochemical properties of cisplatin and related platinum-based drugs, and discusses its uses (either alone or in combination with other drugs) for the treatment of various human cancers. A special attention is paid to its molecular mechanisms of action, and its undesirable side effects.
Collapse
Affiliation(s)
- Shaloam Dasari
- Cellomics and Toxicogenomics Research Laboratory, NIH/NIMHD RCMI-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box 18750, Jackson, MS 39217, USA
| | - Paul Bernard Tchounwou
- Cellomics and Toxicogenomics Research Laboratory, NIH/NIMHD RCMI-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box 18750, Jackson, MS 39217, USA.
| |
Collapse
|
30
|
Zhu J, Chen W, Mi R, Zhou C, Reed N, Höke A. Ethoxyquin prevents chemotherapy-induced neurotoxicity via Hsp90 modulation. Ann Neurol 2014; 74:893-904. [PMID: 23955554 DOI: 10.1002/ana.24004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 07/09/2013] [Accepted: 08/03/2013] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Peripheral neurotoxicity is a major dose-limiting side effect of many chemotherapeutic drugs. Currently there are no effective disease-modifying therapies for chemotherapy-induced peripheral neuropathies, but these side effects of chemotherapy are potentially ideal targets for development of neuroprotective therapies, because candidate drugs can be co- or preadministered before the injury to peripheral axons takes place. METHODS We used a phenotypic drug screening approach to identify ethoxyquin as a potential neuroprotective drug and carried out additional biochemical experiments to identify its mechanism of action. RESULTS We validated the screening results with ethoxyquin and its derivatives and showed that they prevented paclitaxel-induced peripheral neuropathy without blocking paclitaxel's ability to kill tumor cells. Furthermore, we demonstrated that ethoxyquin acts by modulating the chaperone activity of heat shock protein 90 (Hsp90) and blocking the binding of 2 of its client proteins, ataxin-2 and Sf3b2. Ethoxyquin-induced reduction in levels of both of these proteins resulted in prevention of axonal degeneration caused by paclitaxel. INTERPRETATION Ethoxyquin and its novel derivatives as well as other classes of small molecules that act as Hsp90 modulators may offer a new opportunity for development of drugs to prevent chemotherapy-induced axonal degeneration.
Collapse
Affiliation(s)
- Jing Zhu
- Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD
| | | | | | | | | | | |
Collapse
|
31
|
PSY-1, a Taxus chinensis var. mairei Extract, Inhibits Cancer Cell Metastasis by Interfering with MMPs. Nat Prod Commun 2014. [DOI: 10.1177/1934578x1400900228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Cancer is the most common disease worldwide, with death often occurring as a result of metastasis. Thus, interfering with metastasis has been regarded as a promising strategy to improve the current cancer treatments. However, exploration and development of novel anti-metastatic agents remains a major challenge. Recent evidence indicated that a polysaccharide isolated from Taxus yunnanensis suppressed tumor cells proliferation. With the objective of seeking bioactive extracts, we had previously isolated, purified and characterized a complex, water-soluble polysaccharides, PSY-1, from the leaves of Taxus chinensis var. mairei, and identified its anti-neoplastic effects. In this study, we focused on the effects of PSY-1 on cancer metastasis and its mechanism(s). The results illustrated that PSY-1 effectively suppressed the migration and invasion ability of the melanoma cancer cell line B16-F10, caused down-regulation of MMP-2 and MMP-9, and that the NF-κB pathway was involved in the anti-metastatic effects imposed by PSY-1.
Collapse
|
32
|
Tsavaris N, Kosmas C, Skopelitis H, Dimitrakopoulos A, Kopteridis P, Bougas D, Stravodimos K, Mitropoulos D, Alamanis C, Giannopoulos A. Methotrexate-Paclitaxel-Epirubicin-Carboplatin (M-TEC) Combination Chemotherapy in Patients with Advanced Bladder Cancer: An Open Label Phase II Study. J Chemother 2013. [DOI: 10.1179/joc.2005.17.4.444] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
33
|
Zhang XH, Cheng Y, Shin JY, Kim JO, Oh JE, Kang JH. A CDK4/6 inhibitor enhances cytotoxicity of paclitaxel in lung adenocarcinoma cells harboring mutant KRAS as well as wild-type KRAS. Cancer Biol Ther 2013; 14:597-605. [PMID: 23792647 PMCID: PMC3742489 DOI: 10.4161/cbt.24592] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The KRAS gain-of-function mutation confers intrinsic resistance to targeted anti-cancer drugs and cytotoxic chemotherapeutic agents, ultimately leading to treatment failure. KRAS mutation frequency in lung adenocarcinoma is ~15–30%. Novel therapeutic strategies should be developed to improve clinical outcomes in these cases. Deregulation of the p16/cyclin-dependent kinase (CDK) 4/retinoblastoma (Rb) pathway is frequently observed in various cancers and it represents an attractive therapeutic target. We compared the anti-tumor efficacy of genetically knocked-down CDK4 and a pharmacological inhibitor of CDK4/6, CINK4, in KRAS mutation-positive lung adenocarcinoma cells. We also investigated changes in anti-proliferative activity and downstream molecules with these treatments in combination with paclitaxel. CDK4 short interfering RNA (siRNA) significantly increased paclitaxel sensitivity in KRAS mutation-positive H23 cells. CINK4 demonstrated concentration- and time-dependent anti-proliferative activity in 5 adenocarcinoma lines. CINK4 induced G1 arrest by downregulating the p16/cyclin D1/Rb pathway, resulting in apoptotic induction via increased expression of cleaved caspase3, cleaved PARP and Bax. Combined CINK4 and paclitaxel produced synergistic anti-proliferative activity and increased apoptosis through reduced cyclin D1 and Bcl-2 in KRAS mutation-positive cancer cells. These data suggest CDK4 is a promising target for development of anti-cancer drugs and CINK4 combined with paclitaxel may be an effective therapeutic strategy for enhancing anti-tumor efficacy in KRAS mutation-positive lung adenocarcinoma.
Collapse
|
34
|
Choudhury H, Gorain B, Karmakar S, Pal TK. Development and Validation of RP-HPLC Method: Scope of Application in the Determination of Oil Solubility of Paclitaxel. J Chromatogr Sci 2013; 52:68-74. [DOI: 10.1093/chromsci/bms206] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
35
|
Paclitaxel plus platinum or gemcitabine plus platinum in first-line treatment of advanced non-small-cell lung cancer: results from 6 randomized controlled trials. Int J Clin Oncol 2012; 18:1005-13. [DOI: 10.1007/s10147-012-0502-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 11/19/2012] [Indexed: 12/21/2022]
|
36
|
Zhang XH, Shin JY, Kim JO, Oh JE, Yoon SA, Jung CK, Kang JH. Synergistic antitumor efficacy of sequentially combined paclitaxel with sorafenib in vitro and in vivo NSCLC models harboring KRAS or BRAF mutations. Cancer Lett 2012; 322:213-22. [DOI: 10.1016/j.canlet.2012.03.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2011] [Revised: 03/10/2012] [Accepted: 03/12/2012] [Indexed: 12/01/2022]
|
37
|
Du L, Subauste MC, DeSevo C, Zhao Z, Baker M, Borkowski R, Schageman JJ, Greer R, Yang CR, Suraokar M, Wistuba II, Gazdar AF, Minna JD, Pertsemlidis A. miR-337-3p and its targets STAT3 and RAP1A modulate taxane sensitivity in non-small cell lung cancers. PLoS One 2012; 7:e39167. [PMID: 22723956 PMCID: PMC3377607 DOI: 10.1371/journal.pone.0039167] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 05/17/2012] [Indexed: 12/14/2022] Open
Abstract
NSCLC (non-small cell lung cancer) often exhibits resistance to paclitaxel treatment. Identifying the elements regulating paclitaxel response will advance efforts to overcome such resistance in NSCLC therapy. Using in vitro approaches, we demonstrated that over-expression of the microRNA miR-337-3p sensitizes NCI-H1155 cells to paclitaxel, and that miR-337-3p mimic has a general effect on paclitaxel response in NSCLC cell lines, which may provide a novel adjuvant strategy to paclitaxel in the treatment of lung cancer. By combining in vitro and in silico approaches, we identified STAT3 and RAP1A as direct targets that mediate the effect of miR-337-3p on paclitaxel sensitivity. Further investigation showed that miR-337-3p mimic also sensitizes cells to docetaxel, another member of the taxane family, and that STAT3 levels are significantly correlated with taxane resistance in lung cancer cell lines, suggesting that endogenous STAT3 expression is a determinant of intrinsic taxane resistance in lung cancer. The identification of a miR-337-3p as a modulator of cellular response to taxanes, and STAT3 and RAP1A as regulatory targets which mediate that response, defines a novel regulatory pathway modulating paclitaxel sensitivity in lung cancer cells, which may provide novel adjuvant strategies along with paclitaxel in the treatment of lung cancer and may also provide biomarkers for predicting paclitaxel response in NSCLC.
Collapse
Affiliation(s)
- Liqin Du
- Greehey Children's Cancer Research Institute, UT Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Cellular and Structural Biology, UT Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Maria C. Subauste
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Christopher DeSevo
- Division of Basic Sciences, Southwestern Graduate School of Biomedical Sciences, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Zhenze Zhao
- Greehey Children's Cancer Research Institute, UT Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Michael Baker
- Division of Basic Sciences, Southwestern Graduate School of Biomedical Sciences, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Robert Borkowski
- Division of Basic Sciences, Southwestern Graduate School of Biomedical Sciences, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Jeoffrey J. Schageman
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Rachel Greer
- Division of Basic Sciences, Southwestern Graduate School of Biomedical Sciences, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Chin-Rang Yang
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Milind Suraokar
- Department of Pathology, UT MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Ignacio I. Wistuba
- Department of Pathology, UT MD Anderson Cancer Center, Houston, Texas, United States of America
- Department of Thoracic/Head and Neck Medical Oncology, UT MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Adi F. Gazdar
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas, United States of America
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - John D. Minna
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas, United States of America
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Alexander Pertsemlidis
- Greehey Children's Cancer Research Institute, UT Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Cellular and Structural Biology, UT Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Pediatrics, UT Health Science Center at San Antonio, San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|
38
|
Videira M, Almeida AJ, Fabra A. Preclinical evaluation of a pulmonary delivered paclitaxel-loaded lipid nanocarrier antitumor effect. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2011; 8:1208-15. [PMID: 22206945 DOI: 10.1016/j.nano.2011.12.007] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 10/06/2011] [Accepted: 12/15/2011] [Indexed: 12/19/2022]
Abstract
UNLABELLED Lung cancer remains a leading cause of death due to the low efficacy of chemotherapy, mainly related to the administration route used. Therefore, alternative administration routes are needed. Paclitaxel (PTX) is an insoluble anticancer drug active against solid tumors, such as those found in lung cancer, that has stimulated an intense research effort over recent years. Solid lipid nanoparticles (SLNs) are potential carriers for poorly soluble drugs, being biodegradable systems that served as alternatives to the usual colloidal carriers. That system was used to deliver PTX to the lungs and seem to fulfill the requirements for an optimum particulate carrier. Furthermore, PTX-loaded SLN pulmonary administration provided a target administration, which is expected to avoid high concentration of the drug at nontarget tissues, reducing toxicity, and increasing the drug's therapeutic index. The rationale of this study was to deliver a colloidal system to the lung lymphatics through a pulmonary route for cancer therapy. FROM THE CLINICAL EDITOR Paclitaxel-loaded solid lipid nanoparticles were used to target tumors in a murine lung cancer model enabling high PTX concentration in the target with reduced systemic toxicity and increased therapeutic index.
Collapse
Affiliation(s)
- Mafalda Videira
- iMed.UL - Research Institute for Medicines and Pharmaceutical Sciences, School of Pharmacy, Lisbon, Portugal.
| | | | | |
Collapse
|
39
|
Mumenthaler SM, Foo J, Leder K, Choi NC, Agus DB, Pao W, Mallick P, Michor F. Evolutionary modeling of combination treatment strategies to overcome resistance to tyrosine kinase inhibitors in non-small cell lung cancer. Mol Pharm 2011; 8:2069-79. [PMID: 21995722 PMCID: PMC3230244 DOI: 10.1021/mp200270v] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
![]()
Many initially successful anticancer therapies lose effectiveness
over time, and eventually, cancer cells acquire resistance to the
therapy. Acquired resistance remains a major obstacle to improving
remission rates and achieving prolonged disease-free survival. Consequently,
novel approaches to overcome or prevent resistance are of significant
clinical importance. There has been considerable interest in treating
non-small cell lung cancer (NSCLC) with combinations of EGFR-targeted
therapeutics (e.g., erlotinib) and cytotoxic therapeutics (e.g., paclitaxel);
however, acquired resistance to erlotinib, driven by a variety of
mechanisms, remains an obstacle to treatment success. In about 50%
of cases, resistance is due to a T790M point mutation in EGFR, and
T790M-containing cells ultimately dominate the tumor composition and
lead to tumor regrowth. We employed a combined experimental and mathematical
modeling-based approach to identify treatment strategies that impede
the outgrowth of primary T790M-mediated resistance in NSCLC populations.
Our mathematical model predicts the population dynamics of mixtures
of sensitive and resistant cells, thereby describing how the tumor
composition, initial fraction of resistant cells, and degree of selective
pressure influence the time until progression of disease. Model development
relied upon quantitative experimental measurements of cell proliferation
and death using a novel microscopy approach. Using this approach,
we systematically explored the space of combination treatment strategies
and demonstrated that optimally timed sequential strategies yielded
large improvements in survival outcome relative to monotherapies at
the same concentrations. Our investigations revealed regions of the
treatment space in which low-dose sequential combination strategies,
after preclinical validation, may lead to a tumor reduction and improved
survival outcome for patients with T790M-mediated resistance.
Collapse
Affiliation(s)
- Shannon M Mumenthaler
- Center for Applied Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
McTiernan A, Whelan JS. A Phase II Study of Docetaxel for the Treatment of Recurrent Osteosarcoma. Sarcoma 2011; 8:71-6. [PMID: 18521398 PMCID: PMC2395610 DOI: 10.1080/13577140410001711764] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Purpose: To determine the response and toxicity of docetaxel in recurrent osteosarcoma and related spindle cell tumours
of bone. Patients and methods: Fourteen patients, 10 males and four females, were enrolled, median age 30.5 years (range, 17–46).
Diagnosis was: conventional osteosarcoma, 12 patients; periosteal osteosarcoma, one patient; and malignant fibrous
histiocytoma of bone, one patient. Initial chemotherapy had been with doxorubicin and cisplatin in 10 patients, and
multiagent regimens in four. Nine had been treated with second line chemotherapy before receiving docetaxel. Thirteen
patients had lung metastases and one intra-abdominal disease. Docetaxel 100 mg/mα2 was given as a 1-h infusion every
3 weeks. Response was assessed every two cycles to a maximum of six. Results: A total of 43 cycles were given, median of two per patient (range 1–6). Thirteen patients were evaluable for response.
A single partial remission was seen, for a response rate of 8%. Two patients had stable disease, and one patient a
mixed response. Forty cycles were evaluable for toxicity. The principle toxicity was haematological, with a median neutrophil
count of 0.9 (range 0–9.6). There were four episodes of neutropenic sepsis (10%). The only non-haematological toxicity
≥grade 3 was stomatitis, occurring in just one patient. There were no toxic deaths. Conclusion: Docetaxel at this dose and schedule is well tolerated, but is not associated with significant activity in patients
with relapsed osteosarcoma.
Collapse
Affiliation(s)
- Anne McTiernan
- Meyerstein Institute of Oncology Middlesex Hospital UCL Hospitals NHS Trust Mortimer St London W1T 3AA UK
| | | |
Collapse
|
41
|
Gill KK, Nazzal S, Kaddoumi A. Paclitaxel loaded PEG(5000)-DSPE micelles as pulmonary delivery platform: formulation characterization, tissue distribution, plasma pharmacokinetics, and toxicological evaluation. Eur J Pharm Biopharm 2011; 79:276-84. [PMID: 21575719 DOI: 10.1016/j.ejpb.2011.04.017] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 04/27/2011] [Accepted: 04/27/2011] [Indexed: 11/30/2022]
Abstract
The objective of the present study was to evaluate the potential of paclitaxel loaded micelles fabricated from PEG(5000)-DSPE as a sustained release system following pulmonary delivery. PEG(5000)-DSPE micelles containing paclitaxel were prepared by solvent evaporation technique followed by investigation of in vitro release of paclitaxel in lung simulated fluid. Tissue distribution and plasma pharmacokinetics of the PEG-lipid micelles after intratracheal and intravenous administrations were investigated in addition to intratracheally administered taxol. Finally, toxicological profile of PEG(5000)-DSPE was investigated. Paclitaxel was successfully formulated in PEG-lipid micelles with encapsulation efficiency of 95%. The PEG-lipid micelles exhibited a sustained release behavior in the simulated lung fluid. Intratracheally administered polymeric micellar paclitaxel showed highest accumulation of paclitaxel in the lungs with AUC(0-12) in lungs being 45-fold higher than intravenously administered formulation and 3-fold higher than intratracheally delivered taxol. Paclitaxel concentration in other non-targeted tissues and plasma were significantly lower as compared to other groups. Furthermore, toxicity studies showed no significant increase in levels of lung injury markers in PEG(5000)-DSPE treated group as compared to saline-treated group. PEG(5000)-DSPE micelles delivered intratracheally were able to sustain highest paclitaxel concentrations in lungs for long periods of time, thus apprehending their suitability as pulmonary drug carriers.
Collapse
Affiliation(s)
- Kanwaldeep K Gill
- Department of Basic Pharmaceutical Sciences, University of Louisiana at Monroe, Monroe, United States
| | | | | |
Collapse
|
42
|
Joshi S, Tiwari A, Mondal B, Sharma A. Oncoproteomics. Clin Chim Acta 2011; 412:217-26. [DOI: 10.1016/j.cca.2010.10.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Revised: 10/03/2010] [Accepted: 10/03/2010] [Indexed: 11/29/2022]
|
43
|
Wang X, Zheng H, Zhu Z, Wei Y, Chen L. Clinical Pharmacokinetics of Paclitaxel Liposome with a New Route of Administration in Human Based on the Analysis with Ultra Performance Liquid Chromatography. J Pharm Sci 2010; 99:4746-52. [DOI: 10.1002/jps.22169] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
44
|
A Comparison of white and African American outcomes from a three-arm, randomized, phase III multicenter trial of advanced or metastatic non-small cell lung cancer. J Thorac Oncol 2010; 5:993-1000. [PMID: 20593535 DOI: 10.1097/jto.0b013e3181e29cf3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE To investigate the effect of race on the efficacy and safety of standard chemotherapy doublet regimens in African American patients, we conducted a subgroup analysis of a phase III randomized trial. PATIENTS AND METHODS Chemonaïve patients with a performance status of 0 or 1 and stage IIIB or IV non-small cell lung cancer were randomized to arm A: gemcitabine 1000 mg/m2 on days 1 and 8 plus carboplatin area under the curve 5.5 on day 1; arm B: the same schedule of gemcitabine plus paclitaxel 200 mg/m2 on day 1; or arm C: paclitaxel 225 mg/m2 on day 1 plus carboplatin area under the curve 6.0 on day 1. Cycles were repeated every 21 days up to 6. A site selection tool identified institutions with potential to recruit a minority population. Outcome and toxicity data of white and African American patients were compared. RESULTS Of 1135 total patients, 972 were white (85.6%) and 138 were African American (12.2%). Median survival was 8.3 months for white patients (95% confidence interval [CI]: 7.7-9.3) and 9.1 months for African American patients (95% CI: 8.2-11.1). Response rates were 29.1 and 29.0%, respectively. Rates of grade 3 or 4 toxicities were comparable. Among African Americans, median survival was 7.2 months (95% CI: 5.1-10.1) for gemcitabine-carboplatin (n = 47), 10.5 months (95% CI: 7.1-15.4) for gemcitabine-paclitaxel (n = 42), and 10.2 months (95% CI: 8.5-13.2) for paclitaxel-carboplatin (n = 49). CONCLUSION Whites and African Americans had similar outcomes, although there was some variability in survival among African Americans across the three treatment groups.
Collapse
|
45
|
Cheng M, Dong M, Li Y, Sauriol F, Shi QW. A New Taxane with an Opened Oxetane Ring from the Needles of Taxus cuspidata. Chem Biodivers 2010; 7:2035-9. [DOI: 10.1002/cbdv.201000017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
46
|
Wang X, Zhou J, Wang Y, Zhu Z, Lu Y, Wei Y, Chen L. A phase I clinical and pharmacokinetic study of paclitaxel liposome infused in non-small cell lung cancer patients with malignant pleural effusions. Eur J Cancer 2010; 46:1474-80. [DOI: 10.1016/j.ejca.2010.02.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2009] [Revised: 11/16/2009] [Accepted: 02/05/2010] [Indexed: 10/19/2022]
|
47
|
Jauhari S, Singh S, Dash AK. Chapter 7 Paclitaxel. PROFILES OF DRUG SUBSTANCES, EXCIPIENTS, AND RELATED METHODOLOGY 2010; 34:299-344. [PMID: 22469177 DOI: 10.1016/s1871-5125(09)34007-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Saurabh Jauhari
- Department of Pharmacy Sciences, School of Pharmacy and Health Professions, Creighton University, Omaha, NE 68178, USA
| | | | | |
Collapse
|
48
|
Rescue of paclitaxel sensitivity by repression of Prohibitin1 in drug-resistant cancer cells. Proc Natl Acad Sci U S A 2010; 107:2503-8. [PMID: 20133800 DOI: 10.1073/pnas.0910649107] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Paclitaxel has emerged as a front line treatment for aggressive malignancies of the breast, lung, and ovary. Successful therapy of cancer is frequently undermined by the development of paclitaxel resistance. There is a growing need to find other therapeutic targets to facilitate treatment of drug-resistant cancers. Using a proteomics approach, elevated levels of Prohibitin1 (PHB1) and GSTpi were found associated with paclitaxel resistance in discrete subcellular fractions of two drug-resistant sublines relative to their sensitive sublines. Immunofluorescence staining and fractionation studies revealed increased levels of PHB1 on the surface of resistant cell lines. Transiently silencing either PHB1 or GSTpi gene expression using siRNA in the paclitaxel-resistant cancer cell sublines partially sensitized these cells toward paclitaxel. Intriguingly, silencing PHB1 but not GSTpi resulted in activation of the intrinsic apoptosis pathway in response to paclitaxel. Similarly, stably silencing either PHB1 or GSTpi significantly improved paclitaxel sensitivity in A549TR cells both in vitro and in vivo. Our results indicate that PHB1 is a mediator of paclitaxel resistance and that this resistance may depend on the cellular localization of the protein. We suggest PHB1 as a potential target for therapeutic strategies for the treatment of drug-resistant tumors.
Collapse
|
49
|
Treat JA, Gonin R, Socinski MA, Edelman MJ, Catalano RB, Marinucci DM, Ansari R, Gillenwater HH, Rowland KM, Comis RL, Obasaju CK, Belani CP. A randomized, phase III multicenter trial of gemcitabine in combination with carboplatin or paclitaxel versus paclitaxel plus carboplatin in patients with advanced or metastatic non-small-cell lung cancer. Ann Oncol 2009; 21:540-547. [PMID: 19833819 DOI: 10.1093/annonc/mdp352] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Paclitaxel-carboplatin is used as the standard regimen for patients with advanced or metastatic non-small-cell lung cancer (NSCLC). This trial was designed to compare gemcitabine + carboplatin or gemcitabine + paclitaxel to the standard regimen. PATIENTS AND METHODS A total of 1135 chemonaive patients with stage IIIB or IV NSCLC were randomly allocated to receive gemcitabine 1000 mg/m(2) on days 1 and 8 plus carboplatin area under the concentration-time curve (AUC) 5.5 on day 1 (GC), gemcitabine 1000 mg/m(2) on days 1 and 8 plus paclitaxel 200 mg/m(2) on day 1 (GP), or paclitaxel 225 mg/m(2) plus carboplatin AUC 6.0 on day 1 (PC). Stratification was based on disease stage, baseline weight loss, and presence or absence of brain metastases. Cycles were repeated every 21 days for up to six cycles or disease progression. RESULTS Median survival (months) with GC was 7.9 compared with 8.5 for GP and 8.7 for PC. Response rates (RRs) were as follows: GC, 25.3%; GP, 32.1%; and PC, 29.8%. The GC arm was associated with a greater incidence of grade 3 or 4 hematologic events but a lower rate of neurotoxicity and alopecia when compared with GP and PC. CONCLUSIONS Non-platinum and non-paclitaxel gemcitabine-containing doublets demonstrate similar overall survival and RR compared with the standard PC regimen. However, the treatment arms had distinct toxicity profiles.
Collapse
Affiliation(s)
- J A Treat
- US Medical Division, Lilly USA, Indianapolis, IN.
| | | | - M A Socinski
- Division of Hematology/Oncology, Multidisciplinary Thoracic Oncology Group, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - M J Edelman
- Division of Hematology/Oncology, University of Maryland Greenbaum Cancer Center, Baltimore, MD
| | - R B Catalano
- Drexel University College of Medicine, Philadelphia, PA
| | - D M Marinucci
- Drexel University College of Medicine, Philadelphia, PA
| | - R Ansari
- Northern Indiana Cancer Research Consortium, South Bend, IN
| | - H H Gillenwater
- Department of Hematology/Oncology, University of Virginia Cancer Center, Charlottesville, VA
| | - K M Rowland
- Department of Medicine, Carle Clinic Cancer Center, Urbana, IL
| | - R L Comis
- Drexel University College of Medicine, Philadelphia, PA
| | - C K Obasaju
- US Medical Division, Lilly USA, Indianapolis, IN
| | - C P Belani
- Division of Hematology/Oncology, Penn State Hershey Cancer Institute, Hershey, PA, USA
| | | |
Collapse
|
50
|
Langer CJ, Duffy K, Horwitz EM, Litwin S, Rosvold E, Schol J, Keenan E, Nicolaou N, Friedman CD, Ridge JA. Phase I Trial of Concurrent Hyperfractionated Split Course Radiotherapy (HFx RT), Cisplatin (cDDP), and Paclitaxel in Patients with Recurrent, Previously Irradiated, or Treatment-Naïve Locally Advanced Upper Aerodigestive Malignancy. Cancer Invest 2009; 24:164-73. [PMID: 16537186 DOI: 10.1080/07357900500524421] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
PURPOSE Phase I study to determine the maximally tolerated dose (MTD) of cisplatin (cDDP), paclitaxel (P), and concurrent split course hyperfractionated (BID) RT in advanced squamous cell carcinoma of the head and neck (SCCHN) and other upper aerodigestive tumors. MATERIALS AND METHODS Eligibility stipulated ECOG performance status 0-2 and either Tx-naïve, locally advanced, or locally recurrent, previously radiated, surgically unresectable upper aerodigestive cancer. Metastases were permitted if disease was predominantly locoregional. RT-naïve patients received 150 cGy bid x 5 d Q 2 wks x 4. Previously radiated patients received 150 cGy bid x 5, wk 1; then 120 cGy bid x 5 Q 2 wk x 3 (later increased to 150 cGy BID for the entire treatment). Treatment fields included recurrent tumor only with 2 cm margins. Whenever possible, conventional and 3-D conformal techniques were used. Elective nodal radiation was not administered. Starting doses of cDDP and P were 12 mg/m2/d x 5 and 15 mg/m2/d x 5, respectively, Q 2 wk x 4, each given on RT days only. At dose level 2, cDDP was increased to 15 mg/m2/d x 5. At dose level 3, P was increased to 20 mg/m2/d x 5. Granulocyte colony stimulating factor (G-CSF) days 6-12 (off treatment week) was added if cumulative neutropenia precipitated treatment delays. RESULTS Thirty-one patients (21 men, 10 women) were treated. Eight had received prior chemotherapy, 27 prior RT. At dose level three, regular treatment delays of >or=1 week due to slow neutrophil recovery occurred. Addition of G-CSF (dose level 3b) reduced treatment delays from 100 percent to 28 percent and decreased the incidence of Grade >or=2 neutropenia and mucositis. Six of 7 patients at this dose level completed all 4 cycles of treatment and all received full dose RT (60 Gy). No other dose-limiting toxicities occurred. Of 22 assessable patients with locally recurrent SCCHN, 12 (55 percent) responded. Median time to progression in this group was 6 months, with median and one-year survival of 9.5 mos and 41 percent, respectively. CONCLUSION Concurrent daily cisplatin/paclitaxel and split course hyperfractionated RT (60 Gy) is feasible in previously radiated patients. G-CSF, administered between each cycle, reduces the incidence of treatment delays. Activity is promising and toxicity acceptable.
Collapse
Affiliation(s)
- Corey J Langer
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|