1
|
Park SY, Baek YB, Lee CH, Kim HJ, Kim HP, Jeon YJ, Song JE, Jung SB, Kim HJ, Moon KS, Park SI, Lee CM, Kim SH. Establishment of canine mammary gland tumor cell lines harboring PI3K/Akt activation as a therapeutic target. BMC Vet Res 2024; 20:233. [PMID: 38807154 PMCID: PMC11134682 DOI: 10.1186/s12917-024-04085-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 05/16/2024] [Indexed: 05/30/2024] Open
Abstract
Canine mammary gland tumors (MGT) have a poor prognosis in intact female canines, posing a clinical challenge. This study aimed to establish novel canine mammary cancer cell lines from primary tumors and characterize their cellular and molecular features to find potential therapeutic drugs. The MGT cell lines demonstrated rapid cell proliferation and colony formation in an anchorage-independent manner. Vimentin and α-SMA levels were significantly elevated in MGT cell lines compared to normal canine kidney (MDCK) cells, while CDH1 expression was either significantly lower or not detected at all, based on quantitative real-time PCR (qRT-PCR) analysis. Functional annotation and enrichment analysis revealed that epithelial-mesenchymal transition (EMT) phenotypes and tumor-associated pathways, particularly the PI3K/Akt signaling pathway, were upregulated in MGT cells. BYL719 (Alpelisib), a PI3K inhibitor, was also examined for cytotoxicity on the MGT cell lines. The results show that BYL719 can significantly inhibit the proliferation of MGT cell lines in vitro. Overall, our findings suggest that the MGT cell lines may be valuable for future studies on the development, progression, metastasis, and management of tumors.
Collapse
Affiliation(s)
- Seo-Young Park
- Laboratory of Animal Molecular Biochemistry, Department of Animal Science, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Yeong-Bin Baek
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Chonnam National university, Gwangju, 61186, Republic of Korea
| | - Chan-Ho Lee
- Laboratory of Animal Molecular Biochemistry, Department of Animal Science, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyun-Jin Kim
- Laboratory of Animal Molecular Biochemistry, Department of Animal Science, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hwang-Phill Kim
- Department of Molecular Medicine & Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Suwon, 16229, Republic of Korea
| | - Young-Jun Jeon
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jung Eun Song
- Gwangju Animal Medical Center, Gwangju, 62273, Republic of Korea
| | - Su-Bin Jung
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Chonnam National university, Gwangju, 61186, Republic of Korea
| | - Hyo-Jin Kim
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Chonnam National university, Gwangju, 61186, Republic of Korea
| | - Kyeong-Seo Moon
- Laboratory of Veterinary Pathology, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Sang-Ik Park
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Chonnam National university, Gwangju, 61186, Republic of Korea.
| | - Chang-Min Lee
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Sung-Hak Kim
- Laboratory of Animal Molecular Biochemistry, Department of Animal Science, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
2
|
Tripathi AK, Desai PP, Tyagi A, Lampe JB, Srivastava Y, Donkor M, Jones HP, Dzyuba SV, Crossley E, Williams NS, Vishwanatha JK. Short peptides based on the conserved regions of MIEN1 protein exhibit anticancer activity by targeting the MIEN1 signaling pathway. J Biol Chem 2024; 300:105680. [PMID: 38272230 PMCID: PMC10878790 DOI: 10.1016/j.jbc.2024.105680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 01/06/2024] [Accepted: 01/09/2024] [Indexed: 01/27/2024] Open
Abstract
Migration and invasion enhancer 1 (MIEN1) overexpression characterizes several cancers and facilitates cancer cell migration and invasion. Leveraging conserved immunoreceptor tyrosine-based activation motif and prenylation motifs within MIEN1, we identified potent anticancer peptides. Among them, bioactive peptides LA3IK and RP-7 induced pronounced transcriptomic and protein expression changes at sub-IC50 concentrations. The peptides effectively inhibited genes and proteins driving cancer cell migration, invasion, and epithelial-mesenchymal transition pathways, concurrently suppressing epidermal growth factor-induced nuclear factor kappa B nuclear translocation in metastatic breast cancer cells. Specifically, peptides targeted the same signal transduction pathway initiated by MIEN1. Molecular docking and CD spectra indicated the formation of MIEN1-peptide complexes. The third-positioned isoleucine in LA3IK and CVIL motif in RP-7 were crucial for inhibiting breast cancer cell migration. This is evident from the limited migration inhibition observed when MDA-MB-231 cells were treated with scrambled peptides LA3IK SCR and RP-7 SCR. Additionally, LA3IK and RP-7 effectively suppressed tumor growth in an orthotopic breast cancer model. Notably, mice tolerated high intraperitoneal (ip) peptide doses of 90 mg/Kg well, surpassing significantly lower doses of 5 mg/Kg intravenously (iv) and 30 mg/Kg intraperitoneally (ip) used in both in vivo pharmacokinetic studies and orthotopic mouse model assays. D-isomers of LA3IK and RP-7 showed enhanced anticancer activity compared to their L-isomers. D-LA3IK remained stable in mouse plasma for 24 h with 75% remaining, exhibiting superior pharmacokinetic properties over D/L-RP-7. In summary, our findings mark the first report of short peptides based on MIEN1 protein sequence capable of inhibiting cancer signaling pathways, effectively impeding cancer progression both in vitro and in vivo.
Collapse
Affiliation(s)
- Amit K Tripathi
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, USA.
| | - Priyanka P Desai
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Antariksh Tyagi
- Yale Center for Genome Analysis (YCGA), Yale School of Medicine, New Haven, Connecticut, USA
| | - Jana B Lampe
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Yogesh Srivastava
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael Donkor
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Harlan P Jones
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Sergei V Dzyuba
- Department of Chemistry and Biochemistry, Texas Christian University, Fort Worth, Texas, USA
| | - Eric Crossley
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Noelle S Williams
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jamboor K Vishwanatha
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, USA.
| |
Collapse
|
3
|
Sadwal S, Bharati S, Dar ZA, Kaur S. Chemopreventive potential of hydroethanolic Murraya koenigii leaves extract against DMBA induced breast carcinogenesis: In-silico and in-vivo study. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117124. [PMID: 37678421 DOI: 10.1016/j.jep.2023.117124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/28/2023] [Accepted: 09/03/2023] [Indexed: 09/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Murraya koenigii (MK), a member of the Rutaceae family and widely known as the curry-leaf tree, is indigenous to India, Sri Lanka, and other south Asian nations. It is a renowned medicinal herb because of the wide range of bioactive components found in its leaves, such as girinimbine, koenimbine, mahanimbine and mahanine among others. All these bioactive components make this plant beneficial for treating a variety of ailments and diseases. Biological and pharmacological activities of MK include anti-oxidant, anti-microbial, anti-ulcer, anti-helminthic, anti-malarial, anti-trichomonal, hepatoprotective, anti-diabetic, etc. AIM OF THE STUDY: The present study aimed to evaluate the possible protective effect of hydroethanolic Murraya koenigii leaves extract (HEMKLE) against 7,12-Dimethylbenz[a]anthracene (DMBA)-induced breast cancer in rats, which further paves the way for future breast cancer treatment. MATERIALS AND METHODS For the preparation of hydroethanolic Murraya koenigii leaves extract (HEMKLE), Murraya koenigii (MK) leaves were taken from the botanical garden of the Panjab University campus, Chandigarh, and authenticated from the Department of Botany, Panjab University (accession number 22417). The phytochemical characterization of HEMKLE was performed using liquid chromatography-mass spectrometry (LC-MS). Following this, an in-silico molecular docking analysis was performed using Maestro Schrodinger software, and an in-vivo study was conducted. For the in-vivo study, female SD rats were divided into four different groups. Group I (C), Group II (DMBA), Group III (HEMKLE), and Group IV (HEMKLE + DMBA). Histopathogy, oxidative and antioxidant status, immunohistochemistry of estrogen receptor-α, TUNEL assays, mRNA and protein expression of apoptotic pathway genes were conducted in in-vivo study. RESULTS In LC-MS, major phytochemical constituents including flavonoids and carbazole alkaloids were identified. In-silico docking study revealed the strong binding affinity between the identified compounds with caspase-3. Additionally, koenine displayed the highest binding affinity/minimum energy of -9.21 kcal/mol with 6BDV as compared to other phytochemicals. Furthermore, in-vivo experimentation revealed that HEMKLE administration in Group IV(HEMKLE + DMBA) significantly inhibits the tumor incidence and volume as compared to alone DMBA treated group. The antioxidant action of HEMKLE was proven from the in-vivo analysis of antioxidant marker enzymes, histopathology, immunohistochemistry of ER-α studies. Further, increase number of TUNEL positive cells was observed in co-treated animals as compared to alone DMBA treated animals. In Group IV (HEMKLE + DMBA), upregulated expression of pro-apoptotic genes and downregulated expression of anti-apoptotic gene were observed when compared to Group II(DMBA) suggested the apoptotic effect of HEMKLE. CONCLUSION The results of the present study provide clear evidence of the chemopreventive capabilities of HEMKLE in rats with DMBA-induced breast cancer. The observed outcomes could potentially be attributed to the existence of diverse phytochemicals within the HEMKLE.
Collapse
Affiliation(s)
- Shilpa Sadwal
- Department of Biophysics, Panjab University, Chandigarh, India.
| | - Sanjay Bharati
- Manipal College of Health Profession, Manipal Academy of Higher Education, Karnataka, India.
| | - Zahid Ahmad Dar
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India.
| | | |
Collapse
|
4
|
常 馨, 卢 涛, 黄 金. [Preliminary Study on Microvasculature Normalization Induced by Peritumoral Electroacupuncture in Mice With Breast Cancer Xenografts]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2023; 54:972-977. [PMID: 37866955 PMCID: PMC10579062 DOI: 10.12182/20230960401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Indexed: 10/24/2023]
Abstract
Objective To observe the effect of peritumoral electroacupuncture on the induction of vascular normalization in a mouse breast cancer model. Methods A subcutaneous graft model of breast cancer was established with 4T1 breast cancer cell line in female BALB/c mice aged 6-8 weeks. The mice were randomly assigned to three groups, a tumor-bearing group (TG), peritumoral electroacupuncture tumor-bearing group (EATG), and bevacizumab tumor-bearing group (BTG), with 18 mice in each group. The TG mice did not receive any intervention, the EATG mice received peritumoral electroacupuncture for 30 minutes, and the BTG mice were intraperitoneally injected with bevacizumab at 10mg/kg. Immunofluorescence was performed to assess the expression of CD31/alpha smooth muscle actin (α-SMA) and hypoxia-inducible factor 1-alpha (HIF-1α) in the tumor tissue at various points of time, including before intervention and 3 days and 5 days after intervention. Then, 3 days after intervention, observation of morphological changes of the microvessels in the tumor tissue was performed through Hematoxylin and Eosin (HE) staining and scanning electron microscope. Results There was no significant difference in the expression of CD31, α-SMA, and HIF-1α in the tumor tissues of all groups before experimental intervention ( P>0.05). On day 3 of the experimental interventions, the CD31 and HIF-1α expression levels in the tumor tissues of the EATG and BTG mice were significantly reduced ( P<0.01), while α-SMA expression levels were significantly increased ( P<0.01) in both groups. On day 5 of the experimental interventions, the CD31 and HIF-1α expression levels in the tumor tissues of the EATG and BTG mice were still significantly lower than those in the TG mice ( P<0.01), while the α-SMA expression level was significantly higher than that in the TG group ( P<0.05). On day 3 of the experimental interventions, H&E staining showed visible microvessels in the tumor tissues of all 3 groups. In addition, scanning electron microscopic observation showed that the tumor microvessel walls of the TG mice were rough and defective, and that obvious deformities appeared in the lumen. In contrast, the walls of the microvessels of the EATG and BTG mice were generally intact and there was no obvious deformities in the lumen. Conclusion Peritumoral electroacupuncture may induce microvasculature normalization by decreasing microvascular density and increasing pericyte coverage of the neovasculature, thereby improving hypoxic microenvironment of breast cancer in mice.
Collapse
Affiliation(s)
- 馨 常
- 北京中医药大学第三附属医院 (北京 100029)The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing 100029, China
| | - 涛 卢
- 北京中医药大学第三附属医院 (北京 100029)The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing 100029, China
| | - 金昶 黄
- 北京中医药大学第三附属医院 (北京 100029)The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing 100029, China
- 北京中医药大学生命科学学院 (北京 100029)College of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
5
|
Sahu R, Jha S, Pattanayak SP. Therapeutic silencing of mTOR by systemically administered siRNA-loaded neutral liposomal nanoparticles inhibits DMBA-induced mammary carcinogenesis. Br J Cancer 2022; 127:2207-2219. [PMID: 36261586 PMCID: PMC9726943 DOI: 10.1038/s41416-022-02011-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 09/28/2022] [Accepted: 10/04/2022] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Mammary carcinogenesis possesses great challenges due to the lack of effectiveness of the multiple therapeutic options available. Gene therapy-based cancer treatment strategy provides more targeting accuracy, fewer side effects, and higher therapeutic efficiency. Downregulation of the oncogene mTOR by mTOR-siRNA is an encouraging approach to reduce cancer progression. However, its employment as means of therapeutic strategy has been restricted due to the unavailability of a suitable delivery system. METHODS A suitable nanocarrier system made up of 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine (DOPC) has been developed to prevent degradation and for proficient delivery of siRNA. This was followed by in vitro and in vivo anti-breast cancer efficiency analysis of the mTOR siRNA-loaded neutral liposomal formulation (NL-mTOR-siRNA). RESULTS In our experiment, a profound reduction in MCF-7 cell growth, proliferation and invasion was ascertained following extensive downregulation of mTOR expression. NL-mTOR-siRNA suppressed tumour growth and restored morphological alterations of DMBA-induced breast cancer. In addition, neutral liposome enhanced accumulation of siRNA in mammary cancer tissues facilitating its deep cytosolic distribution within the tumour, which allows apoptosis thereby facilitating its anti-tumour potential. CONCLUSION Hence, the current study highlighted the augmented ground for therapies aiming toward cancerous cells to diminish mTOR expression by RNAi in managing mammary carcinoma.
Collapse
Affiliation(s)
- Roja Sahu
- Division of Advanced Pharmacology, Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology (BIT), Mesra, Ranchi, Jharkhand, 835 215, India
| | - Shivesh Jha
- Division of Pharmacognosy and Phytochemistry, Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology (BIT), Mesra, Ranchi, Jharkhand, 835 215, India
| | - Shakti Prasad Pattanayak
- Department of Pharmacy, School of Health Science, Central University of South Bihar (Gaya), Gaya, Bihar, 824 236, India.
| |
Collapse
|
6
|
Seet-Lee C, Yee J, Morahan H, Ross LS, Edwards KM. The effect of aerobic exercise on tumour blood delivery: a systematic review and meta-analysis. Support Care Cancer 2022; 30:8637-8653. [PMID: 35650456 PMCID: PMC9633495 DOI: 10.1007/s00520-022-07132-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 05/09/2022] [Indexed: 01/05/2023]
Abstract
PURPOSE Tumour blood vessels are structurally and functionally abnormal, resulting in areas of hypoxia and heterogeneous blood supply. Aerobic exercise may modulate tumour blood flow and normalise the tumour microenvironment to improve chemotherapy delivery. This systematic review and meta-analysis aimed to evaluate the effect of the aerobic exercise mode on tumour hypoxia, vascularisation and blood flow. METHODS Four online databases were searched. Preclinical and clinical randomised controlled trials examining the effects of aerobic exercise training on hypoxia, vascularisation or blood flow in solid tumours were included. The risk of bias was assessed and a meta-analysis performed. RESULTS Seventeen preclinical studies and one clinical study met criteria. Eleven studies assessed hypoxia, 15 studies assessed vascularisation and seven evaluated blood flow. There was large variability in measurement methods, tumour types and exercise program designs. The overall risk of bias was unclear in clinical and preclinical studies, owing to poor reporting. There was no significant effect of aerobic exercise on hypoxia (SMD = -0.17; 95% CI = -0.62, 0.28; I2 = 60%), vascularisation (SMD = 0.07; 95% CI = -0.40, 0.55; I2 = 71%) or blood flow (SMD = 0.01; 95% CI = -0.59, 0.61; I2 = 63%). CONCLUSION There is heterogeneity in methodology, resulting in evidence that is inconsistent and inconclusive for the effects of aerobic exercise on hypoxia, vascularisation and blood flow. Most evidence of aerobic exercise effects on tumour blood flow is in animal models, with very limited evidence in humans.
Collapse
Affiliation(s)
- Catherine Seet-Lee
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, Australia
| | - Jasmine Yee
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, Australia
- Centre for Medical Psychology & Evidence-Based Decision-Making, School of Psychology, The University of Sydney, Camperdown, Australia
| | - Heidi Morahan
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, Australia
| | - Lois S Ross
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, Australia
- Department for Health, University of Bath, Bath, UK
| | - Kate M Edwards
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, 2006, Australia.
- Charles Perkins Centre, University of Sydney, Camperdown, Australia.
| |
Collapse
|
7
|
Kowalski JP, Pelletier RD, McDonald MG, Kelly EJ, Rettie AE. Pharmacokinetics, metabolism and off-target effects in the rat of 8-[(1H- benzotriazol-1-yl)amino]octanoic acid, a selective inhibitor of human cytochrome P450 4Z1: β-oxidation as a potential augmenting pathway for inhibition. Xenobiotica 2021; 51:901-915. [PMID: 33993844 DOI: 10.1080/00498254.2021.1930281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
8-[(1H-1,2,3-benzotriazol-1-yl)amino]octanoic acid (8-BOA) was recently identified as a selective and potent mechanism-based inactivator (MBI) of breast cancer-associated CYP4Z1 and exhibited favourable inhibitory activity in vitro, thus meriting in vivo characterization.The pharmacokinetics and metabolism of 8-BOA in rats was examined after a single IV bolus dose of 10 mg/kg. A biphasic time-concentration profile resulted in relatively low clearance and a prolonged elimination half-life.The major circulating metabolites identified in plasma were products of β-oxidation; congeners losing two and four methylene groups accounted for >50% of metabolites by peak area. The -(CH2)2 product was characterized previously as a CYP4Z1 MBI and so represents an active metabolite that may contribute to the desired pharmacological effect.Ex vivo analysis of total CYP content in rat liver and kidney microsomes showed that off-target CYP inactivation was minimal; liver microsomal probe substrate metabolism also demonstrated low off-target inactivation. Standard clinical chemistries provided no indication of acute toxicity.In silico simulations using the free concentration of 8-BOA in plasma suggested that the in vivo dose used here may effectively inactivate CYP4Z1 in a xenografted tumour.
Collapse
Affiliation(s)
- John P Kowalski
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA, USA.,Department of Drug Metabolism and Pharmacokinetics, Pfizer Boulder R&D, Boulder, CO, USA
| | - Robert D Pelletier
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA, USA
| | - Matthew G McDonald
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA, USA.,Department of Drug Metabolism and Pharmacokinetics, Pfizer Boulder R&D, Boulder, CO, USA
| | - Edward J Kelly
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, USA
| | - Allan E Rettie
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA, USA
| |
Collapse
|
8
|
Podyacheva EY, Kushnareva EA, Karpov AA, Toropova YG. Analysis of Models of Doxorubicin-Induced Cardiomyopathy in Rats and Mice. A Modern View From the Perspective of the Pathophysiologist and the Clinician. Front Pharmacol 2021; 12:670479. [PMID: 34149423 PMCID: PMC8209419 DOI: 10.3389/fphar.2021.670479] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/20/2021] [Indexed: 12/11/2022] Open
Abstract
Today the pharmacological possibilities of treating cancer are expanding and as a result, life expectancy is increasing against the background of chemotherapy and supportive treatment. In the conditions of successful antitumor treatment, complications associated with its toxic effect on healthy tissues and organs began to come to the fore. Anthracycline cardiomyopathy was the first serious cardiovascular complication to draw the attention of oncologists and cardiologists around the world. Anthracycline drugs such as doxorubicin, epirubicin, idarubicin are still widely used in oncological practice to treat a wide range of solid and hematological malignancies. Doxorubicin-induced cardiomyopathy is closely associated with an increase in oxidative stress, as evidenced by reactive oxygen species (ROS) nduced damage such as lipid peroxidation, and decreased levels of antioxidants. Myofibrillar destruction and dysregulation of intracellular calcium are also important mechanisms, usually associated with doxorubicin-induced cardiotoxicity. Despite the abundance of data on various mechanisms involved in the implementation of doxorubicin-induced cardiotoxicity, a final understanding of the mechanism of the development of doxorubicin cardiomyopathy has not yet been formed. It poses the most significant challenges to the development of new methods of prevention and treatment, as well as to the unambiguous choice of a specific treatment regimen using the existing pharmacological tools. In order to resolve these issues new models that could reflect the development of the chemotherapy drugs effects are needed. In this review we have summarized and analyzed information on the main existing models of doxorubicin cardiomyopathy using small laboratory animals. In addition, this paper discusses further areas of research devoted to the development and validation of new improved models of doxorubicin cardiomyopathy suitable both for studying the mechanisms of its implementation and for the preclinical drugs effectiveness assessment.
Collapse
Affiliation(s)
- Ekaterina Yu Podyacheva
- Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, Saint-Petersburg, Russia
| | - Ekaterina A Kushnareva
- Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, Saint-Petersburg, Russia
| | - Andrei A Karpov
- Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, Saint-Petersburg, Russia
| | - Yana G Toropova
- Almazov National Medical Research Centre, Ministry of Health of the Russian Federation, Saint-Petersburg, Russia
| |
Collapse
|
9
|
Sahu R, Kar RK, Sunita P, Bose P, Kumari P, Bharti S, Srivastava S, Pattanayak SP. LC-MS characterized methanolic extract of zanthoxylum armatum possess anti-breast cancer activity through Nrf2-Keap1 pathway: An in-silico, in-vitro and in-vivo evaluation. JOURNAL OF ETHNOPHARMACOLOGY 2021; 269:113758. [PMID: 33359860 DOI: 10.1016/j.jep.2020.113758] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/26/2020] [Accepted: 12/22/2020] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zanthoxylum armatum DC (Rutaceae) containing flavonoids, alkaloids, coumarins, lignans, amides and terpenoid is well-known for its curative properties against various ailments including cancer. In the current research, phytochemicals present in the methanolic extract of Zanthoxylum armatum bark (MeZb) were characterized by LC-MS/MS analysis and chemotherapeutic potential of this extract was determined on DMBA-induced female Sprague Dawley rats. MATERIALS AND METHODS A simple and fast high-performance liquid chromatography-mass spectroscopy (LC-MS/MS) of MeZb was established followed by in-vitro antioxidant assays. This was followed by in-silico docking analysis as well as cytotoxicity assessment. Successively in-vivo study of MeZb was performed in DMBA-induced Sprague Dawley rats possessing breast cancer along with detailed molecular biology studies involving immunofluorescence, RT-qPCR and Western blot analysis. RESULTS LC-MS/MS investigation revealed the presence of compounds belonging to flavonoid, alkaloid and glycoside groups. MeZb revealed potential antioxidant activity in in-vitro antioxidant assays and strong binding energy of identified compounds was seen from the in-silico study with both HO1 and Keap1 receptor. Furthermore, the antioxidant action of MeZb was proven from the in-vivo analysis of antioxidant marker enzymes (lipid peroxidation, enzymic and non-enzymic antioxidants). This study also revealed upregulation of protective Nrf-2 following downregulation of Keap1 after MeZb treatment with respect to untreated cancerous rats. CONCLUSION These results exhibited anti-breast-cancer potential of MeZb through Nrf2-Keap1 pathway which may be due to the flavonoids, alkaloids and glycosides present in it.
Collapse
Affiliation(s)
- Roja Sahu
- Division of Pharmacology, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835 215, Jharkhand, India
| | - Rajiv Kumar Kar
- Fritz Haber Center for Molecular Dynamics, Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Priyashree Sunita
- Government Pharmacy Institute, Department of Health, Family Welfare and Medical Education, Government of Jharkhand, Bariatu, Ranchi, 834009, India
| | - Pritha Bose
- Division of Pharmacology, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835 215, Jharkhand, India
| | - Puja Kumari
- Division of Pharmacology, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835 215, Jharkhand, India
| | - Salona Bharti
- Division of Pharmacology, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835 215, Jharkhand, India
| | - Sharad Srivastava
- Pharmacognosy & Ethnopharmacology Division, CSIR-National Botanical Research Institute, NBRI-Govt. of India, Lucknow, 226001, India
| | - Shakti P Pattanayak
- Division of Pharmacology, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835 215, Jharkhand, India; Department of Pharmacy, School of Health Sciences, Central University of South Bihar (Gaya), Bihar, 824236, India.
| |
Collapse
|
10
|
Sigdel I, Gupta N, Faizee F, Khare VM, Tiwari AK, Tang Y. Biomimetic Microfluidic Platforms for the Assessment of Breast Cancer Metastasis. Front Bioeng Biotechnol 2021; 9:633671. [PMID: 33777909 PMCID: PMC7992012 DOI: 10.3389/fbioe.2021.633671] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 02/05/2021] [Indexed: 12/27/2022] Open
Abstract
Of around half a million women dying of breast cancer each year, more than 90% die due to metastasis. Models necessary to understand the metastatic process, particularly breast cancer cell extravasation and colonization, are currently limited and urgently needed to develop therapeutic interventions necessary to prevent breast cancer metastasis. Microfluidic approaches aim to reconstitute functional units of organs that cannot be modeled easily in traditional cell culture or animal studies by reproducing vascular networks and parenchyma on a chip in a three-dimensional, physiologically relevant in vitro system. In recent years, microfluidics models utilizing innovative biomaterials and micro-engineering technologies have shown great potential in our effort of mechanistic understanding of the breast cancer metastasis cascade by providing 3D constructs that can mimic in vivo cellular microenvironment and the ability to visualize and monitor cellular interactions in real-time. In this review, we will provide readers with a detailed discussion on the application of the most up-to-date, state-of-the-art microfluidics-based breast cancer models, with a special focus on their application in the engineering approaches to recapitulate the metastasis process, including invasion, intravasation, extravasation, breast cancer metastasis organotropism, and metastasis niche formation.
Collapse
Affiliation(s)
- Indira Sigdel
- Biofluidics Laboratory, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| | - Niraj Gupta
- Biofluidics Laboratory, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| | - Fairuz Faizee
- Biofluidics Laboratory, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| | - Vishwa M Khare
- Eurofins Lancaster Laboratories, Philadelphia, PA, United States
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH, United States
| | - Yuan Tang
- Biofluidics Laboratory, Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH, United States
| |
Collapse
|
11
|
Zhang H, Tang WL, Kheirolomoom A, Fite BZ, Wu B, Lau K, Baikoghli M, Raie MN, Tumbale SK, Foiret J, Ingham ES, Mahakian LM, Tam SM, Cheng RH, Borowsky AD, Ferrara KW. Development of thermosensitive resiquimod-loaded liposomes for enhanced cancer immunotherapy. J Control Release 2021; 330:1080-1094. [PMID: 33189786 PMCID: PMC7906914 DOI: 10.1016/j.jconrel.2020.11.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 11/01/2020] [Accepted: 11/08/2020] [Indexed: 12/12/2022]
Abstract
Resiquimod (R848) is a toll-like receptor 7 and 8 (TLR7/8) agonist with potent antitumor and immunostimulatory activity. However, systemic delivery of R848 is poorly tolerated because of its poor solubility in water and systemic immune activation. In order to address these limitations, we developed an intravenously-injectable formulation with R848 using thermosensitive liposomes (TSLs) as a delivery vehicle. R848 was remotely loaded into TSLs composed of DPPC: DSPC: DSPE-PEG2K (85:10:5, mol%) with 100 mM FeSO4 as the trapping agent inside. The final R848 to lipid ratio of the optimized R848-loaded TSLs (R848-TSLs) was 0.09 (w/w), 10-fold higher than the previously-reported values. R848-TSLs released 80% of R848 within 5 min at 42 °C. These TSLs were then combined with αPD-1, an immune checkpoint inhibitor, and ultrasound-mediated hyperthermia in a neu deletion (NDL) mouse mammary carcinoma model (Her2+, ER/PR negative). Combined with αPD-1, local injection of R848-TSLs showed superior efficacy with complete NDL tumor regression in both treated and abscopal sites achieved in 8 of 11 tumor bearing mice over 100 days. Immunohistochemistry confirmed enhanced CD8+ T cell infiltration and accumulation by R848-TSLs. Systemic delivery of R848-TSLs, combined with local hyperthermia and αPD-1, inhibited tumor growth and extended median survival from 28 days (non-treatment control) to 94 days. Upon re-challenge with reinjection of tumor cells, none of the previously cured mice developed tumors, as compared with 100% of age-matched control mice. The dose of R848 (10 μg for intra-tumoral injection or 6 mg/kg for intravenous injection delivered up to 4 times) was well-tolerated without weight loss or organ hypertrophy. In summary, we developed R848-TSLs that can be administered locally or systematically, resulting in tumor regression and enhanced survival when combined with αPD-1 in mouse models of breast cancer.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA; Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Wei-Lun Tang
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Azadeh Kheirolomoom
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA; Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Brett Z Fite
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Bo Wu
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Kenneth Lau
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Mo Baikoghli
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616, USA
| | - Marina Nura Raie
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Spencer K Tumbale
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Josquin Foiret
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Elizabeth S Ingham
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Lisa M Mahakian
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Sarah M Tam
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - R Holland Cheng
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616, USA
| | | | - Katherine W Ferrara
- Molecular Imaging Program, Department of Radiology, Stanford University, 3165 Porter Drive, Palo Alto, CA 94304, USA.
| |
Collapse
|
12
|
Abstract
Breast cancer is the most common malignancy in women. Basic and translational breast cancer research relies heavily on experimental animal models. Ideally, such models for breast cancer should have commonality with human breast cancer in terms of tumor etiology, biological behavior, pathology, and response to therapeutics. This review introduces current progress in different breast cancer experimental animal models and analyzes their characteristics, advantages, disadvantages, and potential applications. Finally, we propose future research directions for breast cancer animal models.
Collapse
Affiliation(s)
- Li Zeng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Wei Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Ce-Shi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China. E-mail:
| |
Collapse
|
13
|
de Faria Lainetti P, Brandi A, Leis Filho AF, Prado MCM, Kobayashi PE, Laufer-Amorim R, Fonseca-Alves CE. Establishment and Characterization of Canine Mammary Gland Carcinoma Cell Lines With Vasculogenic Mimicry Ability in vitro and in vivo. Front Vet Sci 2020; 7:583874. [PMID: 33195606 PMCID: PMC7655132 DOI: 10.3389/fvets.2020.583874] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/15/2020] [Indexed: 12/11/2022] Open
Abstract
Mammary tumors affect intact and elderly female dogs, and almost 50% of these cases are malignant. Cell culture offers a promising preclinical model to study this disease and creates the opportunity to deposit cell lines at a cell bank to allow greater assay reproducibility and more reliable validation of the results. Another important aspect is the possibility of establishing models and improving our understanding of tumor characteristics, such as vasculogenic mimicry. Because of the importance of cancer cell lines in preclinical models, the present study established and characterized primary cell lines from canine mammary gland tumors. Cell cultures were evaluated for morphology, phenotype, vasculogenic mimicry (VM), and tumorigenicity abilities. We collected 17 primary mammary carcinoma and three metastases and obtained satisfactory results from 10 samples. The cells were transplanted to a xenograft model. All cell lines exhibited a spindle-shaped or polygonal morphology and expressed concomitant pancytokeratin and cytokeratin 8/18. Four cell lines had vasculogenic mimicry ability in vitro, and two cell lines showed in vivo tumorigenicity and VM in the xenotransplanted tumor. Cellular characterization will help create a database to increase our knowledge of mammary carcinomas in dogs, including studies of tumor behavior and the identification of new therapeutic targets.
Collapse
Affiliation(s)
| | - Andressa Brandi
- School of Veterinary Medicine and Animal Science, São Paulo State University-UNESP, Botucatu, Brazil
| | | | | | - Priscila Emiko Kobayashi
- School of Veterinary Medicine and Animal Science, São Paulo State University-UNESP, Botucatu, Brazil
| | - Renée Laufer-Amorim
- School of Veterinary Medicine and Animal Science, São Paulo State University-UNESP, Botucatu, Brazil
| | - Carlos Eduardo Fonseca-Alves
- School of Veterinary Medicine and Animal Science, São Paulo State University-UNESP, Botucatu, Brazil.,Institute of Health Sciences, Universidade Paulista-UNIP, Bauru, Brazil
| |
Collapse
|
14
|
Development and characterization of mammary intraductal (MIND) spontaneous metastasis models for triple-negative breast cancer in syngeneic mice. Sci Rep 2020; 10:4681. [PMID: 32170125 PMCID: PMC7070052 DOI: 10.1038/s41598-020-61679-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 03/02/2020] [Indexed: 02/07/2023] Open
Abstract
Triple-negative breast cancer (TNBC) has a more aggressive phenotype and higher metastasis and recurrence rates than other breast cancer subtypes. TNBC currently lacks a transplantation model that is suitable for clinical simulations of the tumor microenvironment. Intraductal injection of tumor cells into the mammary duct could mimic the occurrence and development of breast cancer. Herein, we injected 4T1 cells into the mammary ducts of BALB/C mice to build a preclinical model of TNBC and optimized the related construction method to observe the occurrence and spontaneous metastasis of tumors. We compared the effects of different cell numbers on tumorigenesis rates, times to tumorigenesis, and metastases to determine the optimal number of cells for modelling. We demonstrated that 4T1-MIND model mice injected with 20,000 cells revealed a suitable tumor formation rate and time, thus indicating a potential treatment time window after distant metastasis. We also injected 20,000 cells directly into the breast fat pad or breast duct for parallel comparison. The results still showed that the 4T1-MIND model provides sufficient treatment time for lung metastases in mice and that it is a more reliable model for early tumor development. The 4T1-MIND model requires continuous improvement and optimization. A suitable and optimized model for translational research and studies on the microenvironment in TNBC should be developed.
Collapse
|
15
|
Cell uptake and anti-tumor effect of liposomes containing encapsulated paclitaxel-bound albumin against breast cancer cells in 2D and 3D cultured models. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2019.101381] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
16
|
Noto FK, Adjan-Steffey V, Tong M, Ravichandran K, Zhang W, Arey A, McClain CB, Ostertag E, Mazhar S, Sangodkar J, DiFeo A, Crawford J, Narla G, Jamling TY. Sprague Dawley Rag2-Null Rats Created from Engineered Spermatogonial Stem Cells Are Immunodeficient and Permissive to Human Xenografts. Mol Cancer Ther 2018; 17:2481-2489. [PMID: 30206106 DOI: 10.1158/1535-7163.mct-18-0156] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 07/10/2018] [Accepted: 09/05/2018] [Indexed: 12/22/2022]
Abstract
The rat is the preferred model for toxicology studies, and it offers distinctive advantages over the mouse as a preclinical research model including larger sample size collection, lower rates of drug clearance, and relative ease of surgical manipulation. An immunodeficient rat would allow for larger tumor size development, prolonged dosing and drug efficacy studies, and preliminary toxicologic testing and pharmacokinetic/pharmacodynamic studies in the same model animal. Here, we created an immunodeficient rat with a functional deletion of the Recombination Activating Gene 2 (Rag2) gene, using genetically modified spermatogonial stem cells (SSC). We targeted the Rag2 gene in rat SSCs with TALENs and transplanted these Rag2-deficient SSCs into sterile recipients. Offspring were genotyped, and a founder with a 27 bp deletion mutation was identified and bred to homozygosity to produce the Sprague-Dawley Rag2 - Rag2 tm1Hera (SDR) knockout rat. We demonstrated that SDR rat lacks mature B and T cells. Furthermore, the SDR rat model was permissive to growth of human glioblastoma cell line subcutaneously resulting in successful growth of tumors. In addition, a human KRAS-mutant non-small cell lung cancer cell line (H358), a patient-derived high-grade serous ovarian cancer cell line (OV81), and a patient-derived recurrent endometrial cancer cell line (OV185) were transplanted subcutaneously to test the ability of the SDR rat to accommodate human xenografts from multiple tissue types. All human cancer cell lines showed efficient tumor uptake and growth kinetics indicating that the SDR rat is a viable host for a range of xenograft studies. Mol Cancer Ther; 17(11); 2481-9. ©2018 AACR.
Collapse
Affiliation(s)
| | | | - Min Tong
- Poseida Therapeutics Inc., San Diego, California
| | | | - Wei Zhang
- Hera BioLabs Inc., Lexington, Kentucky
| | | | | | - Eric Ostertag
- Transposagen Biopharmaceuticals Inc., Lexington, Kentucky
| | - Sahar Mazhar
- Case Western Reserve University, Cleveland, Ohio
| | | | | | - Jack Crawford
- Hera BioLabs Inc., Lexington, Kentucky.,Transposagen Biopharmaceuticals Inc., Lexington, Kentucky
| | - Goutham Narla
- Hera BioLabs Inc., Lexington, Kentucky.,The University of Michigan, Ann Arbor, Michigan
| | - Tseten Y Jamling
- Hera BioLabs Inc., Lexington, Kentucky. .,Transposagen Biopharmaceuticals Inc., Lexington, Kentucky
| |
Collapse
|
17
|
Raccagni I, Belloli S, Valtorta S, Stefano A, Presotto L, Pascali C, Bogni A, Tortoreto M, Zaffaroni N, Daidone MG, Russo G, Bombardieri E, Moresco RM. [18F]FDG and [18F]FLT PET for the evaluation of response to neo-adjuvant chemotherapy in a model of triple negative breast cancer. PLoS One 2018; 13:e0197754. [PMID: 29791503 PMCID: PMC5965848 DOI: 10.1371/journal.pone.0197754] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 05/08/2018] [Indexed: 12/31/2022] Open
Abstract
Rationale Pathological response to neo-adjuvant chemotherapy (NAC) represents a commonly used predictor of survival in triple negative breast cancer (TNBC) and the need to identify markers that predict response to NAC is constantly increasing. Aim of this study was to evaluate the potential usefulness of PET imaging with [18F]FDG and [18F]FLT for the discrimination of TNBC responders to Paclitaxel (PTX) therapy compared to the response assessed by an adapted Response Evaluation Criteria In Solid Tumors (RECIST) criteria based on tumor volume (Tumor Volume Response). Methods Nu/nu mice bearing TNBC lesions of different size were evaluated with [18F]FDG and [18F]FLT PET before and after PTX treatment. SUVmax, Metabolic Tumor Volume (MTV) and Total Lesion Glycolysis (TLG) and Proliferation (TLP) were assessed using a graph-based random walk algorithm. Results We found that in our TNBC model the variation of [18F]FDG and [18F]FLT SUVmax similarly defined tumor response to therapy and that SUVmax variation represented the most accurate parameter. Response evaluation using Tumor Volume Response (TVR) showed that the effectiveness of NAC with PTX was completely independent from lesions size at baseline. Conclusions Our study provided interesting results in terms of sensitivity and specificity of PET in TNBC, revealing the similar performances of [18F]FDG and [18F]FLT in the identification of responders to Paclitaxel.
Collapse
Affiliation(s)
- Isabella Raccagni
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy
- Tecnomed, Foundation of the University of Milano-Bicocca, Monza, Italy
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sara Belloli
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Valtorta
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Medicine and Surgery Department, University of Milano-Bicocca, Monza, Italy
| | - Alessandro Stefano
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy
| | - Luca Presotto
- Nuclear Medicine Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Claudio Pascali
- Nuclear Medicine Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Anna Bogni
- Nuclear Medicine Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Monica Tortoreto
- Molecular Pharmacology Unit, Experimental Oncology and Molecular Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Nadia Zaffaroni
- Molecular Pharmacology Unit, Experimental Oncology and Molecular Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maria Grazia Daidone
- Biomarkers Unit, Experimental Oncology and Molecular Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giorgio Russo
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy
| | | | - Rosa Maria Moresco
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy
- Tecnomed, Foundation of the University of Milano-Bicocca, Monza, Italy
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Medicine and Surgery Department, University of Milano-Bicocca, Monza, Italy
- * E-mail:
| |
Collapse
|
18
|
A Biomimetic Microfluidic Tumor Microenvironment Platform Mimicking the EPR Effect for Rapid Screening of Drug Delivery Systems. Sci Rep 2017; 7:9359. [PMID: 28839211 PMCID: PMC5571192 DOI: 10.1038/s41598-017-09815-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 07/31/2017] [Indexed: 01/18/2023] Open
Abstract
Real-time monitoring of tumor drug delivery in vivo is a daunting challenge due to the heterogeneity and complexity of the tumor microenvironment. In this study, we developed a biomimetic microfluidic tumor microenvironment (bMTM) comprising co-culture of tumor and endothelial cells in a 3D environment. The platform consists of a vascular compartment featuring a network of vessels cultured with endothelial cells forming a complete lumen under shear flow in communication with 3D solid tumors cultured in a tumor compartment. Endothelial cell permeability to both small dye molecules and large liposomal drug carriers were quantified using fluorescence microscopy. Endothelial cell intercellular junction formation was characterized by immunostaining. Endothelial cell permeability significantly increased in the presence of either tumor cell conditioned media (TCM) or tumor cells. The magnitude of this increase in permeability was significantly higher in the presence of metastatic breast tumor cells as compared to non-metastatic ones. Immunostaining revealed impaired endothelial cell-cell junctions in the presence of either metastatic TCM or metastatic tumor cells. Our findings indicate that the bMTM platform mimics the tumor microenvironment including the EPR effect. This platform has a significant potential in applications such as cell-cell/cell-drug carrier interaction studies and rapid screening of cancer drug therapeutics/carriers.
Collapse
|
19
|
Cordeiro YG, Xavier PLP, Rochetti AL, Alexandre PA, Mori CMC, Strefezzi RF, Fukumasu H. Transcriptomic profile reveals molecular events associated to focal adhesion and invasion in canine mammary gland tumour cell lines. Vet Comp Oncol 2017; 16:E89-E98. [DOI: 10.1111/vco.12339] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/20/2017] [Accepted: 06/26/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Y. G. Cordeiro
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, School of Animal Science and Food Engineering; University of São Paulo; Pirassununga Brazil
| | - P. L. P. Xavier
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, School of Animal Science and Food Engineering; University of São Paulo; Pirassununga Brazil
| | - A. L. Rochetti
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, School of Animal Science and Food Engineering; University of São Paulo; Pirassununga Brazil
| | - P. A. Alexandre
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, School of Animal Science and Food Engineering; University of São Paulo; Pirassununga Brazil
| | - C. M. C. Mori
- Department of Pathology, School of Veterinary Medicine and Animal Science; University of São Paulo; São Paulo Brazil
| | - R. F. Strefezzi
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, School of Animal Science and Food Engineering; University of São Paulo; Pirassununga Brazil
| | - H. Fukumasu
- Laboratory of Comparative and Translational Oncology, Department of Veterinary Medicine, School of Animal Science and Food Engineering; University of São Paulo; Pirassununga Brazil
| |
Collapse
|
20
|
Manibusan MK, Touart LW. A comprehensive review of regulatory test methods for endocrine adverse health effects. Crit Rev Toxicol 2017; 47:433-481. [PMID: 28617201 DOI: 10.1080/10408444.2016.1272095] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Development of new endocrine disruption-relevant test methods has been the subject of intensive research efforts for the past several decades, prompted in part by mandates in the 1996 Food Quality Protection Act (FQPA). While scientific understanding and test methods have advanced, questions remain on whether current scientific methods are capable of adequately addressing the complexities of the endocrine system for regulatory health and ecological risk assessments. The specific objective of this article is to perform a comprehensive, detailed evaluation of the adequacy of current test methods to inform regulatory risk assessments of whether a substance has the potential to perturb endocrine-related pathways resulting in human adverse effects. To that end, approximately 42 existing test guidelines (TGs) were considered in the evaluation of coverage for endocrine-related adverse effects. In addition to evaluations of whether test methods are adequate to capture endocrine-related effects, considerations of further enhancements to current test methods, along with the need to develop novel test methods to address existing test method gaps are described. From this specific evaluation, up to 35 test methods are capable of informing whether a chemical substance perturbs known endocrine related biological pathways. Based on these findings, it can be concluded that current validated test methods are adequate to discern substances that may perturb the endocrine system, resulting in an adverse health effect. Together, these test methods predominantly form the core data requirements of a typical food-use pesticide registration submission. It is recognized, however, that the current state of science is rapidly advancing and there is a need to update current test methods to include added enhancements to ensure continued coverage and public health and environmental protection.
Collapse
Affiliation(s)
| | - L W Touart
- b Equiparent Consulting , Woodbridge , VA , USA
| |
Collapse
|
21
|
Chivukula IV, Ramsköld D, Storvall H, Anderberg C, Jin S, Mamaeva V, Sahlgren C, Pietras K, Sandberg R, Lendahl U. Decoding breast cancer tissue-stroma interactions using species-specific sequencing. Breast Cancer Res 2015; 17:109. [PMID: 26265142 PMCID: PMC4534116 DOI: 10.1186/s13058-015-0616-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 07/15/2015] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Decoding transcriptional effects of experimental tissue-tissue or cell-cell interactions is important; for example, to better understand tumor-stroma interactions after transplantation of human cells into mouse (xenografting). Transcriptome analysis of intermixed human and mouse cells has, however, frequently relied on the need to separate the two cell populations prior to transcriptome analysis, which introduces confounding effects on gene expression. METHODS To circumvent this problem, we here describe a bioinformatics-based, genome-wide transcriptome analysis technique, which allows the human and mouse transcriptomes to be decoded from a mixed mouse and human cell population. The technique is based on a bioinformatic separation of the mouse and human transcriptomes from the initial mixed-species transcriptome resulting from sequencing an excised tumor/stroma specimen without prior cell sorting. RESULTS Under stringent separation criteria, i.e., with a read misassignment frequency of 0.2 %, we show that 99 % of the genes can successfully be assigned to be of mouse or human origin, both in silico, in cultured cells and in vivo. We use a new species-specific sequencing technology-referred to as S(3) ("S-cube")-to provide new insights into the Notch downstream response following Notch ligand-stimulation and to explore transcriptional changes following transplantation of two different breast cancer cell lines (luminal MCF7 and basal-type MDA-MB-231) into mammary fat pad tissue in mice of different immunological status. We find that MCF7 and MDA-MB-231 respond differently to fat pad xenografting and the stromal response to transplantation of MCF7 and MDA-MB-231 cells was also distinct. CONCLUSIONS In conclusion, the data show that the S(3) technology allows for faithful recording of transcriptomic changes when human and mouse cells are intermixed and that it can be applied to address a broad spectrum of research questions.
Collapse
Affiliation(s)
- Indira V Chivukula
- Department of Cell and Molecular Biology, Karolinska Institutet, von Eulers väg 3, SE-17177, Stockholm, Sweden.
| | - Daniel Ramsköld
- Department of Cell and Molecular Biology, Karolinska Institutet, von Eulers väg 3, SE-17177, Stockholm, Sweden.
- Ludwig Institute for Cancer Research, Karolinska Institutet, Stockholm, Sweden.
- Present address: Rheumatology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.
| | - Helena Storvall
- Department of Cell and Molecular Biology, Karolinska Institutet, von Eulers väg 3, SE-17177, Stockholm, Sweden.
- Ludwig Institute for Cancer Research, Karolinska Institutet, Stockholm, Sweden.
| | - Charlotte Anderberg
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
- Present address: Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Shaobo Jin
- Department of Cell and Molecular Biology, Karolinska Institutet, von Eulers väg 3, SE-17177, Stockholm, Sweden.
| | - Veronika Mamaeva
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland.
| | - Cecilia Sahlgren
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland.
| | - Kristian Pietras
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
- Present address: Department of Laboratory Medicine, Medicon Village, Lund University, Lund, Sweden.
| | - Rickard Sandberg
- Department of Cell and Molecular Biology, Karolinska Institutet, von Eulers väg 3, SE-17177, Stockholm, Sweden.
- Ludwig Institute for Cancer Research, Karolinska Institutet, Stockholm, Sweden.
| | - Urban Lendahl
- Department of Cell and Molecular Biology, Karolinska Institutet, von Eulers väg 3, SE-17177, Stockholm, Sweden.
| |
Collapse
|
22
|
Spade DJ, McDonnell EV, Heger NE, Sanders JA, Saffarini CM, Gruppuso PA, De Paepe ME, Boekelheide K. Xenotransplantation models to study the effects of toxicants on human fetal tissues. ACTA ACUST UNITED AC 2014; 101:410-22. [PMID: 25477288 DOI: 10.1002/bdrb.21131] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 10/14/2014] [Indexed: 12/11/2022]
Abstract
Many diseases that manifest throughout the lifetime are influenced by factors affecting fetal development. Fetal exposure to xenobiotics, in particular, may influence the development of adult diseases. Established animal models provide systems for characterizing both developmental biology and developmental toxicology. However, animal model systems do not allow researchers to assess the mechanistic effects of toxicants on developing human tissue. Human fetal tissue xenotransplantation models have recently been implemented to provide human-relevant mechanistic data on the many tissue-level functions that may be affected by fetal exposure to toxicants. This review describes the development of human fetal tissue xenotransplant models for testis, prostate, lung, liver, and adipose tissue, aimed at studying the effects of xenobiotics on tissue development, including implications for testicular dysgenesis, prostate disease, lung disease, and metabolic syndrome. The mechanistic data obtained from these models can complement data from epidemiology, traditional animal models, and in vitro studies to quantify the risks of toxicant exposures during human development.
Collapse
Affiliation(s)
- Daniel J Spade
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Contag CH, Lie WR, Bammer MC, Hardy JW, Schmidt TL, Maloney WJ, King BL. Monitoring dynamic interactions between breast cancer cells and human bone tissue in a co-culture model. Mol Imaging Biol 2014; 16:158-66. [PMID: 24008275 DOI: 10.1007/s11307-013-0685-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE Bone is a preferential site of breast cancer metastasis, and models are needed to study this process at the level of the microenvironment. We have used bioluminescence imaging (BLI) and multiplex biomarker immunoassays to monitor dynamic breast cancer cell behaviors in co-culture with human bone tissue. PROCEDURES Femur tissue fragments harvested from hip replacement surgeries were co-cultured with luciferase-positive MDA-MB-231-fLuc cells. BLI was performed to quantify breast cell proliferation and track migration relative to bone tissue. Breast cell colonization of bone tissues was assessed with immunohistochemistry. Biomarkers in co-culture supernatants were profiled with MILLIPLEX(®) immunoassays. RESULTS BLI demonstrated increased MDA-MB-231-fLuc cell proliferation (p < 0.001) in the presence vs. absence of bones and revealed breast cell migration toward bone. Immunohistochemistry illustrated MDA-MB-231-fLuc cell colonization of bone, and MILLIPLEX(®) profiles of culture supernatants suggested breast/bone crosstalk. CONCLUSIONS Breast cell behaviors that facilitate metastasis occur reproducibly in human bone tissue co-cultures and can be monitored and quantified using BLI and multiplex immunoassays.
Collapse
Affiliation(s)
- Christopher H Contag
- Department of Pediatrics, Stanford University School of Medicine, 150E Clark Center, 318 Campus Drive, Stanford, CA, 94305-5427, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Terp MG, Ditzel HJ. Application of proteomics in the study of rodent models of cancer. Proteomics Clin Appl 2014; 8:640-52. [DOI: 10.1002/prca.201300084] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 10/25/2013] [Accepted: 11/27/2013] [Indexed: 01/22/2023]
Affiliation(s)
- Mikkel G. Terp
- Department of Cancer and Inflammation Research; Institute of Molecular Medicine, University of Southern Denmark; Odense Denmark
| | - Henrik J. Ditzel
- Department of Cancer and Inflammation Research; Institute of Molecular Medicine, University of Southern Denmark; Odense Denmark
- Department of Oncology; Odense University Hospital; Odense Denmark
| |
Collapse
|
25
|
Lenfert E, Maenz C, Heinlein C, Jannasch K, Schumacher U, Pantel K, Tolstonog GV, Deppert W, Wegwitz F. Mutant p53 promotes epithelial-mesenchymal plasticity and enhances metastasis in mammary carcinomas of WAP-T mice. Int J Cancer 2014; 136:E521-33. [PMID: 25195563 DOI: 10.1002/ijc.29186] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 07/31/2014] [Accepted: 08/21/2014] [Indexed: 12/20/2022]
Abstract
To study the postulated mutant p53 (mutp53) "gain of function" effects in mammary tumor development, progression and metastasis, we crossed SV40 transgenic WAP-T mice with mutant p53 transgenic WAP-mutp53 mice. Compared to tumors in monotransgenic WAP-T mice, tumors in bitransgenic WAP-T x WAP-mutp53 mice showed higher tumor grading, enhanced vascularization, and significantly increased metastasis. Bitransgenic tumors revealed a gene signature associated with the oncogenic epithelial-mesenchymal transition pathway (EMT gene signature). In cultures of WAP-T tumor-derived G-2 cancer cells, which are comprised of subpopulations displaying "mesenchymal" and "epithelial" phenotypes, this EMT gene signature was associated with the "mesenchymal" compartment. Furthermore, ectopic expression of mutp53 in G-2 cells sufficed to induce a strong EMT phenotype. In contrast to these in vitro effects, monotransgenic and bitransgenic tumors were phenotypically similar suggesting that in vivo the tumor cell phenotype might be under control of the tumor microenvironment. In support, orthotopic transplantation of G-2 cells as well as of G-2 cells expressing ectopic mutp53 into syngeneic mice resulted in tumors with a predominantly epithelial phenotype, closely similar to that of endogenous primary tumors. We conclude that induction of an EMT gene signature by mutp53 in bitransgenic tumors primarily promotes tumor cell plasticity, that is, the probability of tumor cells to undergo EMT processes under appropriate stimuli, thereby possibly increasing their potential to disseminate and metastasize.
Collapse
Affiliation(s)
- Eva Lenfert
- Department for Tumor Biology, University Medical Center Hamburg-Eppendorf (UKE), D-20246, Hamburg, Germany; Department of Tumor Virology, Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, D-20251, Hamburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
El-Abd EA, Sultan AS, Shalaby EA, Matalkah F. Animal Models of Breast Cancer. OMICS APPROACHES IN BREAST CANCER 2014:297-314. [DOI: 10.1007/978-81-322-0843-3_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
27
|
Eccles SA, Aboagye EO, Ali S, Anderson AS, Armes J, Berditchevski F, Blaydes JP, Brennan K, Brown NJ, Bryant HE, Bundred NJ, Burchell JM, Campbell AM, Carroll JS, Clarke RB, Coles CE, Cook GJR, Cox A, Curtin NJ, Dekker LV, dos Santos Silva I, Duffy SW, Easton DF, Eccles DM, Edwards DR, Edwards J, Evans DG, Fenlon DF, Flanagan JM, Foster C, Gallagher WM, Garcia-Closas M, Gee JMW, Gescher AJ, Goh V, Groves AM, Harvey AJ, Harvie M, Hennessy BT, Hiscox S, Holen I, Howell SJ, Howell A, Hubbard G, Hulbert-Williams N, Hunter MS, Jasani B, Jones LJ, Key TJ, Kirwan CC, Kong A, Kunkler IH, Langdon SP, Leach MO, Mann DJ, Marshall JF, Martin LA, Martin SG, Macdougall JE, Miles DW, Miller WR, Morris JR, Moss SM, Mullan P, Natrajan R, O’Connor JPB, O’Connor R, Palmieri C, Pharoah PDP, Rakha EA, Reed E, Robinson SP, Sahai E, Saxton JM, Schmid P, Smalley MJ, Speirs V, Stein R, Stingl J, Streuli CH, Tutt ANJ, Velikova G, Walker RA, Watson CJ, Williams KJ, Young LS, Thompson AM. Critical research gaps and translational priorities for the successful prevention and treatment of breast cancer. Breast Cancer Res 2013; 15:R92. [PMID: 24286369 PMCID: PMC3907091 DOI: 10.1186/bcr3493] [Citation(s) in RCA: 275] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 09/12/2013] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION Breast cancer remains a significant scientific, clinical and societal challenge. This gap analysis has reviewed and critically assessed enduring issues and new challenges emerging from recent research, and proposes strategies for translating solutions into practice. METHODS More than 100 internationally recognised specialist breast cancer scientists, clinicians and healthcare professionals collaborated to address nine thematic areas: genetics, epigenetics and epidemiology; molecular pathology and cell biology; hormonal influences and endocrine therapy; imaging, detection and screening; current/novel therapies and biomarkers; drug resistance; metastasis, angiogenesis, circulating tumour cells, cancer 'stem' cells; risk and prevention; living with and managing breast cancer and its treatment. The groups developed summary papers through an iterative process which, following further appraisal from experts and patients, were melded into this summary account. RESULTS The 10 major gaps identified were: (1) understanding the functions and contextual interactions of genetic and epigenetic changes in normal breast development and during malignant transformation; (2) how to implement sustainable lifestyle changes (diet, exercise and weight) and chemopreventive strategies; (3) the need for tailored screening approaches including clinically actionable tests; (4) enhancing knowledge of molecular drivers behind breast cancer subtypes, progression and metastasis; (5) understanding the molecular mechanisms of tumour heterogeneity, dormancy, de novo or acquired resistance and how to target key nodes in these dynamic processes; (6) developing validated markers for chemosensitivity and radiosensitivity; (7) understanding the optimal duration, sequencing and rational combinations of treatment for improved personalised therapy; (8) validating multimodality imaging biomarkers for minimally invasive diagnosis and monitoring of responses in primary and metastatic disease; (9) developing interventions and support to improve the survivorship experience; (10) a continuing need for clinical material for translational research derived from normal breast, blood, primary, relapsed, metastatic and drug-resistant cancers with expert bioinformatics support to maximise its utility. The proposed infrastructural enablers include enhanced resources to support clinically relevant in vitro and in vivo tumour models; improved access to appropriate, fully annotated clinical samples; extended biomarker discovery, validation and standardisation; and facilitated cross-discipline working. CONCLUSIONS With resources to conduct further high-quality targeted research focusing on the gaps identified, increased knowledge translating into improved clinical care should be achievable within five years.
Collapse
Affiliation(s)
- Suzanne A Eccles
- The Institute of Cancer Research, 15 Cotswold Road, London SM2 5MG, UK
| | - Eric O Aboagye
- Imperial College London, Exhibition Rd, London SW7 2AZ, UK
| | - Simak Ali
- Imperial College London, Exhibition Rd, London SW7 2AZ, UK
| | | | - Jo Armes
- Kings College London, Strand, London WC2R 2LS, UK
| | | | - Jeremy P Blaydes
- University of Southampton, University Road, Southampton SO17 1BJ, UK
| | - Keith Brennan
- University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Nicola J Brown
- University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Helen E Bryant
- University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Nigel J Bundred
- University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | | | | | - Jason S Carroll
- Cancer Research UK, Cambridge Research Institute/University of Cambridge, Trinity Lane, Cambridge CB2 1TN, UK
| | - Robert B Clarke
- University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Charlotte E Coles
- Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge CB2 0QQ, UK
| | - Gary JR Cook
- Kings College London, Strand, London WC2R 2LS, UK
| | - Angela Cox
- University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Nicola J Curtin
- Newcastle University, Claremont Road, Newcastle upon Tyne NE1 7RU, UK
| | | | | | - Stephen W Duffy
- Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Douglas F Easton
- Cancer Research UK, Cambridge Research Institute/University of Cambridge, Trinity Lane, Cambridge CB2 1TN, UK
| | - Diana M Eccles
- University of Southampton, University Road, Southampton SO17 1BJ, UK
| | - Dylan R Edwards
- University of East Anglia, Earlham Road, Norwich NR4 7TJ, UK
| | - Joanne Edwards
- University of Glasgow, University Avenue, Glasgow G12 8QQ, UK
| | - D Gareth Evans
- University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Deborah F Fenlon
- University of Southampton, University Road, Southampton SO17 1BJ, UK
| | | | - Claire Foster
- University of Southampton, University Road, Southampton SO17 1BJ, UK
| | | | | | - Julia M W Gee
- University of Cardiff, Park Place, Cardiff CF10 3AT, UK
| | - Andy J Gescher
- University of Leicester, University Road, Leicester LE1 4RH, UK
| | - Vicky Goh
- Kings College London, Strand, London WC2R 2LS, UK
| | - Ashley M Groves
- University College London, Gower Street, London WC1E 6BT, UK
| | | | - Michelle Harvie
- University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Bryan T Hennessy
- Royal College of Surgeons Ireland, 123, St Stephen’s Green, Dublin 2, Ireland
| | | | - Ingunn Holen
- University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Sacha J Howell
- University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Anthony Howell
- University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | | | | | | | - Bharat Jasani
- University of Cardiff, Park Place, Cardiff CF10 3AT, UK
| | - Louise J Jones
- Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Timothy J Key
- University of Oxford, Wellington Square, Oxford OX1 2JD, UK
| | - Cliona C Kirwan
- University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Anthony Kong
- University of Oxford, Wellington Square, Oxford OX1 2JD, UK
| | - Ian H Kunkler
- University of Edinburgh, South Bridge, Edinburgh EH8 9YL, UK
| | - Simon P Langdon
- University of Edinburgh, South Bridge, Edinburgh EH8 9YL, UK
| | - Martin O Leach
- The Institute of Cancer Research, 15 Cotswold Road, London SM2 5MG, UK
| | - David J Mann
- Imperial College London, Exhibition Rd, London SW7 2AZ, UK
| | - John F Marshall
- Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Lesley Ann Martin
- The Institute of Cancer Research, 15 Cotswold Road, London SM2 5MG, UK
| | - Stewart G Martin
- University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | | | | | | | | | - Sue M Moss
- Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Paul Mullan
- Queen’s University Belfast, University Road, Belfast BT7 1NN, UK
| | - Rachel Natrajan
- The Institute of Cancer Research, 15 Cotswold Road, London SM2 5MG, UK
| | | | | | - Carlo Palmieri
- The University of Liverpool, Brownlow Hill, Liverpool L69 7ZX, UK
| | - Paul D P Pharoah
- Cancer Research UK, Cambridge Research Institute/University of Cambridge, Trinity Lane, Cambridge CB2 1TN, UK
| | - Emad A Rakha
- University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Elizabeth Reed
- Princess Alice Hospice, West End Lane, Esher KT10 8NA, UK
| | - Simon P Robinson
- The Institute of Cancer Research, 15 Cotswold Road, London SM2 5MG, UK
| | - Erik Sahai
- London Research Institute, 44 Lincoln’s Inn Fields, London WC2A 3LY, UK
| | - John M Saxton
- University of East Anglia, Earlham Road, Norwich NR4 7TJ, UK
| | - Peter Schmid
- Brighton and Sussex Medical School, University of Sussex, Brighton, East Sussex BN1 9PX, UK
| | | | | | - Robert Stein
- University College London, Gower Street, London WC1E 6BT, UK
| | - John Stingl
- Cancer Research UK, Cambridge Research Institute/University of Cambridge, Trinity Lane, Cambridge CB2 1TN, UK
| | | | | | | | | | - Christine J Watson
- Cancer Research UK, Cambridge Research Institute/University of Cambridge, Trinity Lane, Cambridge CB2 1TN, UK
| | - Kaye J Williams
- University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Leonie S Young
- Royal College of Surgeons Ireland, 123, St Stephen’s Green, Dublin 2, Ireland
| | | |
Collapse
|
28
|
Mousseau Y, Mollard S, Faucher-Durand K, Richard L, Nizou A, Cook-Moreau J, Baaj Y, Qiu H, Plainard X, Fourcade L, Funalot B, Sturtz FG. Fingolimod potentiates the effects of sunitinib malate in a rat breast cancer model. Breast Cancer Res Treat 2011; 134:31-40. [PMID: 22160641 DOI: 10.1007/s10549-011-1903-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 11/24/2011] [Indexed: 02/04/2023]
Abstract
Most of the antiangiogenic strategies used in oncology principally target endothelial cells through the vascular endothelial growth factor (VEGF) pathway. Multiple kinase inhibitors can secondarily reduce mural cell stabilization of the vessels by blocking platelet-derived growth factor receptor (PDGFR) activity. However, sphingosine-1-phosphate (S1P), which is also implicated in mural cell recruitment, has yet to be targeted in clinical practice. We therefore investigated the potential of a simultaneous blockade of the PDGF and S1P pathways on the chemotactic responses of vascular smooth muscle cells (VSMCs) and the resulting effects of this blockade on breast tumor growth. Due to crosstalk between the S1P and PDGF pathways, we used AG1296 and/or VPC-23019 to inhibit PDGFR-β and S1PR1/S1PR3 receptors, respectively. We showed that S1PR1 and S1PR3 are the principal receptors that mediate the S1P chemotactic signal on rat VSMCs and that they act synergistically with PDGFR-β during PDGF-B signaling. We also showed that simultaneous blockade of the PDGFR-β and S1PR1/S1PR3 signals had a synergistic effect, decreasing VSMC migration velocity toward endothelial cell and breast carcinoma cell-secreted cytokines by 65-90%. This blockade also strongly decreased the ability of VSMCs to form a three-dimensional cell network. Similar results were obtained with the combination of sunitinib malate (a VEGFR/PDGFR kinase inhibitor) and fingolimod (an S1P analog). Sunitinib malate is a clinically approved cancer treatment, whereas fingolimod is currently indicated only for treatment of multiple sclerosis. Orally administered, the combination of these drugs greatly decreased rat breast tumor growth in a syngeneic cancer model (Walker 256). This bi-therapy did not exert cumulative toxicity and histological analysis of the tumors revealed normalization of the tumor vasculature. The simultaneous blockade of these signaling pathways with sunitinib malate and fingolimod may provide an effective means of reducing tumor angiogenesis, and may improve the delivery of other chemotherapies.
Collapse
MESH Headings
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Aorta, Thoracic/pathology
- Carcinoma 256, Walker/blood supply
- Carcinoma 256, Walker/drug therapy
- Carcinoma 256, Walker/pathology
- Cell Movement
- Cells, Cultured
- Drug Screening Assays, Antitumor
- Drug Synergism
- Female
- Fingolimod Hydrochloride
- Indoles/administration & dosage
- Male
- Mammary Neoplasms, Experimental/blood supply
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/physiology
- Neoplasm Transplantation
- Propylene Glycols/administration & dosage
- Proto-Oncogene Proteins c-sis/pharmacology
- Proto-Oncogene Proteins c-sis/physiology
- Pyrroles/administration & dosage
- Rats
- Rats, Sprague-Dawley
- Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Receptors, Lysosphingolipid/metabolism
- Receptors, Lysosphingolipid/physiology
- Sphingosine/administration & dosage
- Sphingosine/analogs & derivatives
- Sphingosine-1-Phosphate Receptors
- Statistics, Nonparametric
- Sunitinib
- Tumor Burden/drug effects
- Tyrphostins/pharmacology
Collapse
Affiliation(s)
- Yoanne Mousseau
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Limoges, Limoges, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|