51
|
Ross C, Salmon A, Strong R, Fernandez E, Javors M, Richardson A, Tardif S. Metabolic consequences of long-term rapamycin exposure on common marmoset monkeys (Callithrix jacchus). Aging (Albany NY) 2016; 7:964-73. [PMID: 26568298 PMCID: PMC4694066 DOI: 10.18632/aging.100843] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Rapamycin has been shown to extend lifespan in rodent models, but the effects on metabolic health and function have been widely debated in both clinical and translational trials. Prior to rapamycin being used as a treatment to extend both lifespan and healthspan in the human population, it is vital to assess the side effects of the treatment on metabolic pathways in animal model systems, including a closely related non-human primate model. In this study, we found that long-term treatment of marmoset monkeys with orally-administered encapsulated rapamycin resulted in no overall effects on body weight and only a small decrease in fat mass over the first few months of treatment. Rapamycin treated subjects showed no overall changes in daily activity counts, blood lipids, or significant changes in glucose metabolism including oral glucose tolerance. Adipose tissue displayed no differences in gene expression of metabolic markers following treatment, while liver tissue exhibited suppressed G6Pase activity with increased PCK and GPI activity. Overall, the marmosets revealed only minor metabolic consequences of chronic treatment with rapamycin and this adds to the growing body of literature that suggests that chronic and/or intermittent rapamycin treatment results in improved health span and metabolic functioning. The marmosets offer an interesting alternative animal model for future intervention testing and translational modeling.
Collapse
Affiliation(s)
- Corinna Ross
- Department of Arts & Sciences, Texas A&M University San Antonio, San Antonio, TX 78224, USA.,Barshop Institute for Longevity & Aging Studies, University of Texas Health Science Center San Antonio, San Antonio, TX 78224, USA
| | - Adam Salmon
- Barshop Institute for Longevity & Aging Studies, University of Texas Health Science Center San Antonio, San Antonio, TX 78224, USA.,Geriatric Research, Education & Clinical Center, South Texas Veteran's Health Care System, San Antonio, TX 78224, USA
| | - Randy Strong
- Barshop Institute for Longevity & Aging Studies, University of Texas Health Science Center San Antonio, San Antonio, TX 78224, USA
| | - Elizabeth Fernandez
- Barshop Institute for Longevity & Aging Studies, University of Texas Health Science Center San Antonio, San Antonio, TX 78224, USA.,Geriatric Research, Education & Clinical Center, South Texas Veteran's Health Care System, San Antonio, TX 78224, USA
| | - Marty Javors
- Barshop Institute for Longevity & Aging Studies, University of Texas Health Science Center San Antonio, San Antonio, TX 78224, USA.,Geriatric Research, Education & Clinical Center, South Texas Veteran's Health Care System, San Antonio, TX 78224, USA
| | - Arlan Richardson
- University of Oklahoma Health Sciences Center and the Oklahoma City VA Medical Center, Oklahoma City, OK 73104, USA
| | - Suzette Tardif
- Barshop Institute for Longevity & Aging Studies, University of Texas Health Science Center San Antonio, San Antonio, TX 78224, USA.,Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78224, USA
| |
Collapse
|
52
|
Response to: Tyrosine 397 phosphorylation is critical for FAK-promoted Rac1 activation and invasive properties in oral squamous cell carcinoma cells. J Transl Med 2016; 96:1027. [PMID: 27562307 DOI: 10.1038/labinvest.2016.81] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
53
|
Newman JC, Milman S, Hashmi SK, Austad SN, Kirkland JL, Halter JB, Barzilai N. Strategies and Challenges in Clinical Trials Targeting Human Aging. J Gerontol A Biol Sci Med Sci 2016; 71:1424-1434. [PMID: 27535968 PMCID: PMC5055653 DOI: 10.1093/gerona/glw149] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 07/12/2016] [Indexed: 12/21/2022] Open
Abstract
Interventions that target fundamental aging processes have the potential to transform human health and health care. A variety of candidate drugs have emerged from basic and translational research that may target aging processes. Some of these drugs are already in clinical use for other purposes, such as metformin and rapamycin. However, designing clinical trials to test interventions that target the aging process poses a unique set of challenges. This paper summarizes the outcomes of an international meeting co-ordinated by the NIH-funded Geroscience Network to further the goal of developing a translational pipeline to move candidate compounds through clinical trials and ultimately into use. We review the evidence that some drugs already in clinical use may target fundamental aging processes. We discuss the design principles of clinical trials to test such interventions in humans, including study populations, interventions, and outcomes. As examples, we offer several scenarios for potential clinical trials centered on the concepts of health span (delayed multimorbidity and functional decline) and resilience (response to or recovery from an acute health stress). Finally, we describe how this discussion helped inform the design of the proposed Targeting Aging with Metformin study.
Collapse
Affiliation(s)
- John C Newman
- Division of Geriatrics, University of California San Francisco
| | - Sofiya Milman
- Department of Medicine, Division of Endocrinology and.,Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York
| | - Shahrukh K Hashmi
- Department of Hematology and Transplant Center, Mayo Clinic, Rochester, Minnesota
| | - Steve N Austad
- Department of Biology, University of Alabama at Birmingham
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota
| | - Jeffrey B Halter
- Geriatrics Center and Institute of Gerontology, University of Michigan, Ann Arbor
| | - Nir Barzilai
- Department of Medicine, Division of Endocrinology and .,Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
54
|
Goldman JW, Mendenhall MA, Rettinger SR. Hyperglycemia Associated With Targeted Oncologic Treatment: Mechanisms and Management. Oncologist 2016; 21:1326-1336. [PMID: 27473045 DOI: 10.1634/theoncologist.2015-0519] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 06/09/2016] [Indexed: 12/15/2022] Open
Abstract
: Molecularly targeted cancer therapy has rapidly changed the landscape of oncologic care, often improving patients' prognosis without causing as substantial a quality-of-life decrement as cytotoxic chemotherapy does. Nevertheless, targeted agents can cause side effects that may be less familiar to medical oncologists and that require the attention and expertise of subspecialists. In this review, we focus on hyperglycemia, which can occur with use of new anticancer agents that interact with cell proliferation pathways. Key mediators of these pathways include the tyrosine kinase receptors insulin growth factor receptor 1 (IGF-1R) and epidermal growth factor receptor (EGFR), as well as intracellular signaling molecules phosphatidylinositol 3-kinase (PI3K), AKT, and mammalian target of rapamycin (mTOR). We summarize available information on hyperglycemia associated with agents that inhibit these molecules within the larger context of adverse event profiles. The highest incidence of hyperglycemia is observed with inhibition of IGF-1R or mTOR, and although the incidence is lower with PI3K, AKT, and EGFR inhibitors, hyperglycemia is still a common adverse event. Given the interrelationships between the IGF-1R and cell proliferation pathways, it is important for oncologists to understand the etiology of hyperglycemia caused by anticancer agents that target those pathways. We also discuss monitoring and management approaches for treatment-related hyperglycemia for some of these agents, with a focus on our experience during the clinical development of the EGFR inhibitor rociletinib. IMPLICATIONS FOR PRACTICE Treatment-related hyperglycemia is associated with several anticancer agents. Many cancer patients may also have preexisting or undiagnosed diabetes or glucose intolerance. Screening can identify patients at risk for hyperglycemia before treatment with these agents. Proper monitoring and management of symptoms, including lifestyle changes and pharmacologic intervention, may allow patients to continue benefiting from use of anticancer agents.
Collapse
Affiliation(s)
- Jonathan W Goldman
- Division of Hematology and Oncology, David Geffen School of Medicine at UCLA, Santa Monica, California, USA
| | - Melody A Mendenhall
- Division of Hematology and Oncology, David Geffen School of Medicine at UCLA, Santa Monica, California, USA
| | - Sarah R Rettinger
- Endocrinology Medical Group of Orange County, Inc., Orange, California, USA
| |
Collapse
|
55
|
Prediction of response to everolimus in neuroendocrine tumors: evaluation of clinical, biological and histological factors. Invest New Drugs 2016; 34:654-62. [PMID: 27230034 DOI: 10.1007/s10637-016-0363-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 05/18/2016] [Indexed: 12/19/2022]
Abstract
Objectives Several targeted therapies are available for metastatic neuroendocrine tumours (NETs) but no predictive factor of response to these treatments has been identified yet. Our aim was to identify and evaluate clinical, biological, histological and functional markers of response to everolimus. Methods We retrospectively reviewed 53 patients with NETs treated with everolimus (68 % in clinical trials). Clinical, biological and histological data were analyzed. The functional marker p-p70S6K, a main effector of the mTOR pathway, was studied by immunohistochemistry in 43 cases. Prognostic factors of progression-free survival (PFS) were studied by Kaplan Meier analysis. Results All patients had metastatic and progressive disease before everolimus treatment. Objective response was 9 % and median PFS was 8.1 (4.7-11.5) months. Hypercholesterolemia (HR = 0.13, p < 0.0001) was associated with longer PFS, whereas presence of bone metastases (HR = 3.1, p < 0.001) and overexpression of p-p70S6K by tumor cells (HR = 2.5, p = 0.01) were associated with shorter PFS under everolimus at multivariate analysis. Conclusion Clinical markers are not useful to predict response to everolimus. However, occurrence of hypercholesterolemia under treatment may be an early marker of response. Prospective studies are required to confirm these results and to assess whether p-p70S6K immunostaining is a prognostic or predictive marker of no-response to everolimus.
Collapse
|
56
|
Grote S, Almstedt HC, Tarleton HP. Cardiometabolic Health Among Cancer Survivors: A 13-Week Pilot Study of a Combined Aerobic and Resistance Training Program. Oncol Nurs Forum 2016; 43:306-15. [PMID: 27105192 DOI: 10.1188/16.onf.306-315] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PURPOSE/OBJECTIVES To explore the feasibility of combined aerobic and resistance training (CART) as a safe method of improving cardiometabolic health among cancer survivors.
. DESIGN Descriptive and longitudinal pilot study for exercise intervention.
. SETTING University campus in Los Angeles, California.
. SAMPLE A multiethnic population of cancer survivors (N = 11) was recruited by convenience sampling and physician referral.
. METHODS Consenting participants were prescribed CART for one hour per day, three days per week for 13 weeks.
. MAIN RESEARCH VARIABLES Components of cardiometabolic health were measured, including resting heart rate (HRrest), blood pressure, body mass index, waist circumference, body fat percentage, and android fat percentage at baseline and after 13 weeks of training. Fasting blood glucose, insulin, adiponectin, leptin, tumor necrosis factor alpha, and C-reactive protein (CRP) also were assessed at baseline and after 13 weeks of training.
. FINDINGS More than half of the participants reported living with at least two other chronic diseases or conditions in addition to a cancer diagnosis. Five of six African American and Hispanic participants reported the presence of at least two risk factors for metabolic syndrome, compared to one of five Caucasian participants. After 13 weeks of training, participants experienced an average decrease in waist circumference. Decrease in waist circumference was associated with a decrease in CRP. A relationship also was suggested between number of exercise sessions attended and improvement in HRrest.
. CONCLUSIONS A CART intervention among cancer survivors should continue to be explored in a larger sample to establish efficacy and effectiveness at improving cardiometabolic health. Because of the higher risk of comorbidity among cancer survivors in comparison to cancer-free adults, improving cardiometabolic health is as important as monitoring cancer recurrence. A need exists for increased attention to the post-treatment cardiometabolic health of cancer survivors and also for examining potential cardiometabolic health disparities among non-Caucasian cancer survivors.
. IMPLICATIONS FOR NURSING CART may be a plausible alternative to reduce the risk of metabolic syndrome and improve cardiometabolic health among cancer survivors. Additional studies that continue to explore the efficacy and effectiveness of CART may provide more information to help nurses and physicians determine whether the cancer survivorship care plan should include an exercise-based alternative to intervene on cardiometabolic health.
Collapse
|
57
|
García-Jiménez C, Gutiérrez-Salmerón M, Chocarro-Calvo A, García-Martinez JM, Castaño A, De la Vieja A. From obesity to diabetes and cancer: epidemiological links and role of therapies. Br J Cancer 2016; 114:716-22. [PMID: 26908326 PMCID: PMC4984860 DOI: 10.1038/bjc.2016.37] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/14/2016] [Accepted: 01/22/2016] [Indexed: 12/13/2022] Open
Abstract
Increasing evidence suggests a complex relationship between obesity, diabetes and cancer. Here we review the evidence for the association between obesity and diabetes and a wide range of cancer types. In many cases the evidence for a positive association is strong, but for other cancer types a more complex picture emerges with some site-specific cancers associated with obesity but not to diabetes, and some associated with type I but not type II diabetes. The evidence therefore suggests the existence of cumulative common and differential mechanisms influencing the relationship between these diseases. Importantly, we highlight the influence of antidiabetics on cancer and antineoplastic agents on diabetes and in particular that antineoplastic targeting of insulin/IGF-1 signalling induces hyperglycaemia that often evolves to overt diabetes. Overall, a coincidence of diabetes and cancer worsens outcome and increases mortality. Future epidemiology should consider dose and time of exposure to both disease and treatment, and should classify cancers by their molecular signatures. Well-controlled studies on the development of diabetes upon cancer treatment are necessary and should identify the underlying mechanisms responsible for these reciprocal interactions. Given the global epidemic of diabetes, preventing both cancer occurrence in diabetics and the onset of diabetes in cancer patients will translate into a substantial socioeconomic benefit.
Collapse
Affiliation(s)
- Custodia García-Jiménez
- Department of Basic Health Sciences, Faculty of Health Science, University Rey Juan Carlos, 28922 Alcorcon, Madrid, Spain
| | - María Gutiérrez-Salmerón
- Department of Basic Health Sciences, Faculty of Health Science, University Rey Juan Carlos, 28922 Alcorcon, Madrid, Spain
| | - Ana Chocarro-Calvo
- Department of Basic Health Sciences, Faculty of Health Science, University Rey Juan Carlos, 28922 Alcorcon, Madrid, Spain
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford, UK
| | - Jose Manuel García-Martinez
- Department of Basic Health Sciences, Faculty of Health Science, University Rey Juan Carlos, 28922 Alcorcon, Madrid, Spain
| | - Angel Castaño
- Department of Basic Health Sciences, Faculty of Health Science, University Rey Juan Carlos, 28922 Alcorcon, Madrid, Spain
- Department of Pathology, Hospital Universitario de Fuenlabrada, 28942 Madrid, Spain
| | - Antonio De la Vieja
- Endocrine Tumour Unit (UFIEC), Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain
| |
Collapse
|
58
|
Nabors LB, Surboeck B, Grisold W. Complications from pharmacotherapy. HANDBOOK OF CLINICAL NEUROLOGY 2016; 134:235-250. [PMID: 26948358 DOI: 10.1016/b978-0-12-802997-8.00014-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The pharmacotherapy management of cancers of the nervous system has significant overlap with systemic solid cancers that may utilize similar drugs or agents. There is however a unique aspect related to central nervous system (CNS) cancers where therapies directed against a malignant process may have enhanced toxicities or toxicities unique to the CNS. In addition, many agents used to treat CNS malignancies have unique CNS toxicities that may require a specific intervention. This chapter attempts to review conventional and biologic therapies utilized for CNS malignancies and characterize expected and, if known, unique toxicities.
Collapse
Affiliation(s)
- L Burt Nabors
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Birgit Surboeck
- Department of Neurology, Kaiser-Franz-Josef Hospital, Vienna, Austria
| | - Wolfgang Grisold
- Department of Neurology, Kaiser-Franz-Josef Hospital, Vienna, Austria; Medical University of Vienna, Vienna, Austria
| |
Collapse
|
59
|
Complications of hyperglycaemia with PI3K-AKT-mTOR inhibitors in patients with advanced solid tumours on Phase I clinical trials. Br J Cancer 2015; 113:1541-7. [PMID: 26554652 PMCID: PMC4705886 DOI: 10.1038/bjc.2015.373] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 09/16/2015] [Accepted: 10/05/2015] [Indexed: 01/17/2023] Open
Abstract
Background: PI3K–AKT–mTOR inhibitors (PAMi) are promising anticancer treatments. Hyperglycaemia is a mechanism-based toxicity of these agents and is becoming increasingly important with their use in larger numbers of patients. Methods: Retrospective case-control study comparing incidence and severity of hyperglycaemia (all grades) between a case group of 387 patients treated on 18 phase I clinical trials with PAMi (78 patients with PI3Ki, 138 with mTORi, 144 with AKTi and 27 with PI3K/mTORi) and a control group of 109 patients treated on 10 phase I clinical trials with agents not directly targeting the PAM pathway. Diabetic patients were excluded in both groups. Results: The incidence of hyperglycaemia was not significantly different between cases and controls (86.6% vs 80.7%, respectively, P=0.129). However, high grade (grade 3–4) hyperglycaemia was more frequent in the PAMi group than in controls (6.7% vs 0%, respectively, P=0.005). The incidence of grade 3–4 hyperglycaemia was greater with AKT and multikinase inhibitors compared with other PAMi (P<0.001). All patients with high-grade hyperglycaemia received antihyperglycemic treatment and none developed severe metabolic complications (diabetic ketoacidosis or hyperosmolar hyperglycemic nonketotic state). High-grade hyperglycaemia was the cause of permanent PAMi discontinuation in nine patients. Conclusions: PI3K–AKT–mTOR inhibitors are associated with small (6.7%) but statistically significant increased risk of high-grade hyperglycaemia compared with non-PAM targeting agents. However, PAMi-induced hyperglycaemia was not found to be associated with severe metabolic complications in this non-diabetic population of patients with advanced cancers.
Collapse
|
60
|
Abstract
The mammalian target of rapamycin (mTOR) inhibitors are drugs, primarily used as immunosuppressors that are now frequently used as antineoplastic therapies in various cancers (such as advanced renal cell carcinoma, advanced breast cancer, progressive pancreatic neuroendocrine tumors). They act on mTOR signaling pathway which plays a key role in regulating cell growth as well as lipid and glucose metabolism. Treatment with mTOR inhibitors is associated with a high incidence of hyperglycemia and new-onset diabetes, ranging from 13% to 50% in the clinical trials in which they have been used as anticancer therapies. The rate of severe hyperglycemia is also increased, ranging from 4 to 12% in the main phase III clinical trials. Due to limited human studies, the pathophysiology of mTOR inhibitor-induced hyperglycemia has not yet been totally clarified. However, data from animal studies suggest that the mechanisms responsible for hyperglycemia with mTOR inhibitors are likely due to the combination of impaired insulin secretion and insulin resistance. Due to the high rate of hyperglycemia associated with the use of mTOR inhibitors, a close and personalized follow-up of blood glucose is recommended in all patients.
Collapse
Affiliation(s)
- Bruno Vergès
- Endocrinology-Diabetology Department, University-Hospital, and, Medicine University, Dijon, France; INSERM CRI 866, Medicine University, Dijon, France; Faculté de Médecine, Université de Nantes, Nantes, France.
| | - Bertrand Cariou
- INSERM CRI 866, Medicine University, Dijon, France; Clinique d'Endocrinologie, l'institut du thorax, CHU de Nantes, Nantes, France; INSERM, UMR1087, l'institut du thorax, Nantes, France; Faculté de Médecine, Université de Nantes, Nantes, France
| |
Collapse
|
61
|
Saifullah B, Hussein MZB. Inorganic nanolayers: structure, preparation, and biomedical applications. Int J Nanomedicine 2015; 10:5609-33. [PMID: 26366081 PMCID: PMC4562743 DOI: 10.2147/ijn.s72330] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hydrotalcite-like compounds are two-dimensional inorganic nanolayers also known as clay minerals or anionic clays or layered double hydroxides/layered hydroxy salts, and have emerged as a single type of material with numerous biomedical applications, such as drug delivery, gene delivery, cosmetics, and biosensing. Inorganic nanolayers are promising materials due to their fascinating properties, such as ease of preparation, ability to intercalate different type of anions (inorganic, organic, biomolecules, and even genes), high thermal stability, delivery of intercalated anions in a sustained manner, high biocompatibility, and easy biodegradation. Inorganic nanolayers have been the focus for researchers over the last decade, resulting in widening application horizons, especially in the field of biomedical science. These nanolayers have been widely applied in drug and gene delivery. They have also been applied in biosensing technology, and most recently in bioimaging science. The suitability of inorganic nanolayers for application in drug delivery, gene delivery, biosensing technology, and bioimaging science makes them ideal materials to be applied for theranostic purposes. In this paper, we review the structure, methods of preparation, and latest advances made by inorganic nanolayers in such biomedical applications as drug delivery, gene delivery, biosensing, and bioimaging.
Collapse
Affiliation(s)
- Bullo Saifullah
- Materials Synthesis and Characterization Laboratory, Institute of Advanced Technology (ITMA), Universiti Putra Malaysia, Serdang, Malaysia
| | - Mohd Zobir B Hussein
- Materials Synthesis and Characterization Laboratory, Institute of Advanced Technology (ITMA), Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
62
|
Yin F, Yang C, Wang Q, Zeng S, Hu R, Lin G, Tian J, Hu S, Lan RF, Yoon HS, Lu F, Wang K, Yong KT. A Light-Driven Therapy of Pancreatic Adenocarcinoma Using Gold Nanorods-Based Nanocarriers for Co-Delivery of Doxorubicin and siRNA. Theranostics 2015; 5:818-33. [PMID: 26000055 PMCID: PMC4440440 DOI: 10.7150/thno.11335] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 03/14/2015] [Indexed: 01/05/2023] Open
Abstract
In this work, we report the engineering of polyelectrolyte polymers coated Gold nanorods (AuNRs)-based nanocarriers that are capable of co-delivering small interfering RNA (siRNA) and an anticancer drug doxorubicin (DOX) to Panc-1 cancer cells for combination of both chemo- and siRNA-mediated mutant K-Ras gene silencing therapy. Superior anticancer efficacy was observed through synergistic combination of promoted siRNA and DOX release upon irradiating the nanoplex formulation with 665 nm light. Our antitumor study shows that the synergistic effect of AuNRs nanoplex formulation with 665 nm light treatment is able to inhibit the in vivo tumor volume growth rate by 90%. The antitumor effect is contributed from the inactivation of K-Ras gene and thereby causing a profound synthesis (S) phase arrest in treated Panc-1 cells. Our study shows that the percentage of Panc-1 cells treated by nanoplex formulation with S phase is determined to be 35% and it is 17% much higher than that of Panc-1 cells without any treatments. The developed nanotherapy formulation here, that combines chemotherapy, RNA silencing and NIR window light-mediated therapy, will be seen to be the next natural step to be taken in the clinical research for improving the therapeutic outcomes of the pancreatic adenocarcinoma treatment.
Collapse
Affiliation(s)
- Feng Yin
- 1. School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Chengbin Yang
- 1. School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Qianqian Wang
- 3. Laboratory of Chemical Genetics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Shuwen Zeng
- 1. School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore 639798, Singapore
- 6. CINTRA CNRS/NTU/THALES, UMI 3288, Research Techno Plaza, 50 Nanyang Drive, Border X Block, Singapore, 637553
| | - Rui Hu
- 1. School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Guimiao Lin
- 5. The key lab of Biomedical Engineering and Research Institute of Uropoiesis and Reproduction, School of Medical Sciences, Shenzhen University, Shenzhen, 518060, China
| | - Jinglin Tian
- 5. The key lab of Biomedical Engineering and Research Institute of Uropoiesis and Reproduction, School of Medical Sciences, Shenzhen University, Shenzhen, 518060, China
| | - Siyi Hu
- 7. School of Science, Changchun University of Science and Technology, Changchun, 130022, China
| | - Rong Feng Lan
- 8. Institute of Research and Continuing Education, Hong Kong Baptist University (Shenzhen), Shenzhen 518057, China
| | - Ho Sup Yoon
- 2. Division of Structural Biology & Biochemistry, School of Biological Sciences, Nanyang Technological University, Singapore 639798, Singapore
- 9. Department of Genetic Engineering, College of Life Sciences, Kyung Hee University, Yongin-si Gyeonggi-do, 446-701, Republic of Korea
| | - Fei Lu
- 3. Laboratory of Chemical Genetics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Kuan Wang
- 4. Nanomedicine Program and Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei 115, Taiwan
| | - Ken-Tye Yong
- 1. School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore 639798, Singapore
| |
Collapse
|
63
|
Dembitz V, Lalic H, Ostojic A, Vrhovac R, Banfic H, Visnjic D. The mechanism of synergistic effects of arsenic trioxide and rapamycin in acute myeloid leukemia cell lines lacking typical t(15;17) translocation. Int J Hematol 2015; 102:12-24. [PMID: 25758096 DOI: 10.1007/s12185-015-1776-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 02/27/2015] [Accepted: 03/02/2015] [Indexed: 11/28/2022]
Abstract
Arsenic trioxide (ATO) has potent clinical activity in the treatment of patients with acute promyelocytic leukemia (APL), but is much less efficacious in acute myeloid leukemia (AML) lacking t(15;17) translocation. Recent studies have indicated that the addition of mammalian target of rapamycin (mTOR) inhibitors may increase the sensitivity of malignant cells to ATO. The aim of the present study was to test for possible synergistic effects of ATO and rapamycin at therapeutically achievable doses in non-APL AML cells. In HL-60 and U937 cell lines, the inhibitory effects of low concentrations of ATO and rapamycin were synergistic and more pronounced in U937 cells. The combination of drugs increased apoptosis in HL-60 cells and increased the percentage of cells in G(0)/G(1) phase in both cell lines. In U937 cells, rapamycin alone increased the activity of mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) and the addition of ATO decreased the level of phosphorylated ERK, Ser473 phosphorylated Akt and anti-apoptotic Mcl-1 protein. Primary AML cells show high sensitivity to growth-inhibitory effects of rapamycin alone or in combination with ATO. The results of the present study reveal the mechanism of the synergistic effects of two drugs at therapeutically achievable doses in non-APL AML cells.
Collapse
Affiliation(s)
- Vilma Dembitz
- Department of Physiology and Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Salata 3, POB 978, 10 000, Zagreb, Croatia
| | | | | | | | | | | |
Collapse
|
64
|
Abstract
There is increasing use of tyrosine kinase inhibitors as targeted therapy for several malignancies. Sunitinib is the first-line treatment for renal cancer and we report a case of a man receiving this medication who also had diabetes. When started on sunitinib he experienced improvement in his diabetes control with reduction in his insulin requirements, which later worsened when sunitinib was reduced or stopped. Several retrospective studies have been performed demonstrating this effect with sunitinib, but to date no prospective studies have been reported. Most tyrosine kinase inhibitors reduce blood glucose levels in diabetics, but some agents, such as nilotinib, may increase them. There is no consensus on the mechanism of action of sunitinib in reducing glucose levels. Several theories have been postulated, such as increased insulin secretion, increased insulin sensitivity, reduced loss of islet cells, the gastrointestinal side effects of sunitinib, or an interaction with other antihyperglycaemic agents.
Collapse
Affiliation(s)
| | - Thinn Pwint
- Department of Oncology, Buckinghamshire NHS Trust, Aylesbury, Buckinghamshire, UK
| |
Collapse
|
65
|
Marques RB, Aghai A, de Ridder CMA, Stuurman D, Hoeben S, Boer A, Ellston RP, Barry ST, Davies BR, Trapman J, van Weerden WM. High Efficacy of Combination Therapy Using PI3K/AKT Inhibitors with Androgen Deprivation in Prostate Cancer Preclinical Models. Eur Urol 2014; 67:1177-1185. [PMID: 25220373 DOI: 10.1016/j.eururo.2014.08.053] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 08/20/2014] [Indexed: 12/27/2022]
Abstract
BACKGROUND The phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/AKT pathway is frequently activated during prostate cancer (PCa) progression through loss or mutation of the phosphatase and tensin homolog (PTEN) gene. Following the androgen receptor (AR) pathway, it is the second major driver of PCa growth. OBJECTIVE To assess efficacy of novel PI3K/AKT-targeted therapies in PCa models, as a single agent and in combination with androgen deprivation. DESIGN, SETTING, AND PARTICIPANTS Twelve human PCa cell lines were tested in vitro for sensitivity to the AKT inhibitor AZD5363 and the PI3K beta/delta inhibitor AZD8186. The combination of AZD5363 and AZD8186 with castration was evaluated in vivo in PTEN-negative versus PTEN-positive patient-derived xenografts. Tumors and plasma were collected for biomarker analysis. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS In vitro growth inhibition was determined by methylthiazolyldiphenyl-tetrazolium bromide assay. In vivo efficacy was monitored by caliper measurements of subcutaneous tumor volume. PI3K/AKT and AR pathway activity was analyzed by Western blot, enzyme-linked immunosorbent assay, and real-time polymerase chain reaction. RESULTS AND LIMITATIONS AZD5363 and AZD8186 inhibited in vitro growth of 10 of 12 and 7 of 12 PCa cell lines, respectively, with increased sensitivity under androgen depletion. In vivo, AZD5363 and AZD8186 as single agents significantly inhibited growth of PTEN-negative PC346C xenografts compared to placebo by 60% and 66%, respectively. Importantly, combination of either agent with castration resulted in long-lasting tumor regression, which persisted after treatment cessation. Expression of AR-target genes kallikrein-related peptidase 3 (KLK3, also known as PSA); transmembrane protease, serine 2 (TMPRSS2); and FK506 binding protein 5 (FKBP5) was upregulated after PI3K/AKT inhibition. Neither compound inhibited tumor growth in the PTEN-positive PC310 model. CONCLUSIONS Combination with hormonal therapy improved efficacy of PI3K/AKT-targeted agents in PTEN-negative PCa models. Upregulation of AR-target genes upon PI3K/AKT inhibition suggests a compensatory crosstalk between the PI3K-AR pathways. These data strongly advocate for further clinical evaluation. PATIENT SUMMARY Inactivation of the PTEN gene is a common event promoting prostate cancer (PCa) progression. This preclinical study illustrates the potent anticancer activity of novel PTEN-targeted drugs on PCa models, particularly in combination with hormonal therapy.
Collapse
Affiliation(s)
- Rute B Marques
- Department of Urology, Erasmus MC, University Medical Center Rotterdam, The Netherlands.
| | - Ashraf Aghai
- Department of Urology, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Corrina M A de Ridder
- Department of Urology, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Debra Stuurman
- Department of Urology, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Sander Hoeben
- Department of Urology, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Agnes Boer
- Department of Urology, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | | | - Simon T Barry
- Oncology Innovative Medicines, AstraZeneca, Macclesfield, UK
| | - Barry R Davies
- Oncology Innovative Medicines, AstraZeneca, Macclesfield, UK
| | - Jan Trapman
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Wytske M van Weerden
- Department of Urology, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| |
Collapse
|