1
|
Marin JJG, Serrano MA, Herraez E, Lozano E, Ortiz-Rivero S, Perez-Silva L, Reviejo M, Briz O. Impact of genetic variants in the solute carrier ( SLC) genes encoding drug uptake transporters on the response to anticancer chemotherapy. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:27. [PMID: 39143954 PMCID: PMC11322974 DOI: 10.20517/cdr.2024.42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 08/16/2024]
Abstract
Cancer drug resistance constitutes a severe limitation for the satisfactory outcome of these patients. This is a complex problem due to the co-existence in cancer cells of multiple and synergistic mechanisms of chemoresistance (MOC). These mechanisms are accounted for by the expression of a set of genes included in the so-called resistome, whose effectiveness often leads to a lack of response to pharmacological treatment. Additionally, genetic variants affecting these genes further increase the complexity of the question. This review focuses on a set of genes encoding members of the transportome involved in drug uptake, which have been classified into the MOC-1A subgroup of the resistome. These proteins belong to the solute carrier (SLC) superfamily. More precisely, we have considered here several members of families SLC2, SLC7, SLC19, SLC22, SLCO, SLC28, SLC29, SLC31, SLC46, and SLC47 due to the impact of their expression and genetic variants in anticancer drug uptake by tumor cells or, in some cases, general bioavailability. Changes in their expression levels and the appearance of genetic variants can contribute to the Darwinian selection of more resistant clones and, hence, to the development of a more malignant phenotype. Accordingly, to address this issue in future personalized medicine, it is necessary to characterize both changes in resistome genes that can affect their function. It is also essential to consider the time-dependent dimension of these features, as the genetic expression and the appearance of genetic variants can change during tumor progression and in response to treatment.
Collapse
Affiliation(s)
- Jose J. G. Marin
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid 28029, Spain
| | - Maria A. Serrano
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid 28029, Spain
| | - Elisa Herraez
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid 28029, Spain
| | - Elisa Lozano
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid 28029, Spain
| | - Sara Ortiz-Rivero
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid 28029, Spain
| | - Laura Perez-Silva
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
| | - Maria Reviejo
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid 28029, Spain
| | - Oscar Briz
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, Institute for Biomedical Research of Salamanca (IBSAL), Salamanca 37007, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid 28029, Spain
| |
Collapse
|
2
|
Wylaź M, Kaczmarska A, Pajor D, Hryniewicki M, Gil D, Dulińska-Litewka J. Exploring the role of PI3K/AKT/mTOR inhibitors in hormone-related cancers: A focus on breast and prostate cancer. Biomed Pharmacother 2023; 168:115676. [PMID: 37832401 DOI: 10.1016/j.biopha.2023.115676] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/06/2023] [Accepted: 10/06/2023] [Indexed: 10/15/2023] Open
Abstract
Breast cancer (BC) and prostate cancer (PC) are at the top of the list when it comes to the most common types of cancers worldwide. The phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathway is important, in that it strongly influences the development and progression of these tumors. Previous studies have emphasized the key role of inhibitors of the PIK3/AKT/mTOR signaling pathway in the treatment of BC and PC, and it remains to be a crucial method of treatment. In this review, the inhibitors of these signaling pathways are compared, as well as their effectiveness in therapy and potential as therapeutic agents. The use of these inhibitors as polytherapy is evaluated, especially with the use of hormonal therapy, which has shown promising results.
Collapse
Affiliation(s)
- Mateusz Wylaź
- Student Scientific Group at Jagiellonian University Medical College, Faculty of Medicine, Medical Biochemistry, ul. Mikołaja Kopernika Street 7C, 31-034 Krakow, Poland
| | - Anna Kaczmarska
- Student Scientific Group at Jagiellonian University Medical College, Faculty of Medicine, Medical Biochemistry, ul. Mikołaja Kopernika Street 7C, 31-034 Krakow, Poland
| | - Dawid Pajor
- Student Scientific Group at Jagiellonian University Medical College, Faculty of Medicine, Medical Biochemistry, ul. Mikołaja Kopernika Street 7C, 31-034 Krakow, Poland
| | - Matthew Hryniewicki
- Student Scientific Group at Jagiellonian University Medical College, Faculty of Medicine, Medical Biochemistry, ul. Mikołaja Kopernika Street 7C, 31-034 Krakow, Poland
| | - Dorota Gil
- Chair of Medical Biochemistry, Jagiellonian University Medical College, ul. Mikołaja Kopernika Street 7C, 31-034 Krakow, Poland
| | - Joanna Dulińska-Litewka
- Chair of Medical Biochemistry, Jagiellonian University Medical College, ul. Mikołaja Kopernika Street 7C, 31-034 Krakow, Poland.
| |
Collapse
|
3
|
Skorda A, Lauridsen AR, Wu C, Huang J, Mrackova M, Winther NI, Jank V, Sztupinszki Z, Strauss R, Bilgin M, Maeda K, Liu B, Luo Y, Jäättelä M, Kallunki T. Activation of invasion by oncogenic reprogramming of cholesterol metabolism via increased NPC1 expression and macropinocytosis. Oncogene 2023; 42:2495-2506. [PMID: 37420029 PMCID: PMC10421736 DOI: 10.1038/s41388-023-02771-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 06/22/2023] [Accepted: 06/27/2023] [Indexed: 07/09/2023]
Abstract
Cancer cells are dependent on cholesterol, and they possess strictly controlled cholesterol homeostasis mechanisms. These allow them to smoothly switch between cholesterol synthesis and uptake to fulfill their needs and to adapt environmental changes. Here we describe a mechanism of how cancer cells employ oncogenic growth factor signaling to promote uptake and utilization of extracellular cholesterol via Myeloid Zinc Finger 1 (MZF1)-mediated Niemann Pick C1 (NPC1) expression and upregulated macropinocytosis. Expression of p95ErbB2, highly oncogenic, standard-treatment resistant form of ErbB2 mobilizes lysosomes and activates EGFR, invasion and macropinocytosis. This is connected to a metabolic shift from cholesterol synthesis to uptake due to macropinocytosis-enabled flow of extracellular cholesterol. NPC1 increase facilitates extracellular cholesterol uptake and is necessary for the invasion of ErbB2 expressing breast cancer spheroids and ovarian cancer organoids, indicating a regulatory role for NPC1 in the process. The ability to obtain cholesterol as a byproduct of increased macropinocytosis allows cancer cells to direct the resources needed for the energy-consuming cholesterol synthesis towards other activities such as invasion. These results demonstrate that macropinocytosis is not only an alternative energy source for cancer cells but also an efficient way to provide building material, such as cholesterol, for its macromolecules and membranes.
Collapse
Affiliation(s)
- Aikaterini Skorda
- Cancer Invasion and Resistance, Danish Cancer Institute, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Anna Røssberg Lauridsen
- Cancer Invasion and Resistance, Danish Cancer Institute, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Chengnan Wu
- Cancer Invasion and Resistance, Danish Cancer Institute, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Jinrong Huang
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Monika Mrackova
- Cancer Invasion and Resistance, Danish Cancer Institute, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Nuggi Ingholt Winther
- Cancer Invasion and Resistance, Danish Cancer Institute, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Vanessa Jank
- Cancer Invasion and Resistance, Danish Cancer Institute, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Zsofia Sztupinszki
- Translational Cancer Genomics, Danish Cancer Institute, Copenhagen, Denmark
| | - Robert Strauss
- Genome Integrity Group, Danish Cancer Institute, Copenhagen, Denmark
| | - Mesut Bilgin
- Lipidomics Core Facility, Danish Cancer Institute, Copenhagen, Denmark
| | - Kenji Maeda
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Institute, Copenhagen, Denmark
| | - Bin Liu
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Institute, Copenhagen, Denmark
| | - Yonglun Luo
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Marja Jäättelä
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Institute, Copenhagen, Denmark
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tuula Kallunki
- Cancer Invasion and Resistance, Danish Cancer Institute, Strandboulevarden 49, 2100, Copenhagen, Denmark.
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
4
|
Jiang H, Li M, Du K, Ma C, Cheng Y, Wang S, Nie X, Fu C, He Y. Traditional Chinese Medicine for adjuvant treatment of breast cancer: Taohong Siwu Decoction. Chin Med 2021; 16:129. [PMID: 34857023 PMCID: PMC8638166 DOI: 10.1186/s13020-021-00539-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/17/2021] [Indexed: 02/06/2023] Open
Abstract
The high incidence of breast cancer is the greastest threat to women' health all over the world. Among them, HER-2 positive breast cancer has the characteristics of high malignancy, easy recurrence and metastasis, and poor prognosis. Traditional Chinese medicine (TCM) has a rich theoretical basis and clinical application for breast cancer. TCM believes that blood stasis syndrome is one of the important pathogenesis of breast formation and development. Taohong Siwu Decoction (TSHWD) is based on the "First Prescription of Gynecology" Siwu Decoction. It is widely used in various blood stasis and blood deficiency syndromes, mainly in gynecological blood stasis. Clinical studies have found that THSWD can treat breast cancer by reducing blood vessel and lymphangiogenesis with auxiliary chemotherapy. In this study, we aim to explore the material basis and mechanism of THSWD in the treatment of HER-2 positive breast cancer through literature review and network pharmacology studies. Through a literature review of the traditional application, chemical composition of Chinese herbal medicine of THSWD, as well as its clinical reports and pharmacological research on breast cancer treatment. Meanwhile, we conducted "component-pathway-target" network through network pharmacology reveals the main material basis, possible targets and pathways of THSWD in inhibiting HER-2 positive breast cancer. Literature review and network pharmacology research results had predicted that, baicalein, kaempferol, caffeic acid, amygdalin, quercetin, ferulic acid, gallic acid, catalpol, hydroxysafflor yellow A, paeoniflorin in THSWD are the main effective chemical composition. THSWD regulates 386 protein targets and 166 pathways related to breast cancer. The molecular mechanism is mainly to improve the microenvironment of tumor cells, regulate the process of tumor cell EMT, and inhibit tumor cell proliferation and metastasis. This study revealed the mechanism of action of THSWD in the treatment of HER-2 positive breast cancer through literature review and network pharmacology studies, providing a scientific basis for clinical application.
Collapse
Affiliation(s)
- Huajuan Jiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Minmin Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Kequn Du
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Chuan Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Yanfen Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Shengju Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Xin Nie
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Chaomei Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Yao He
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, 611137, Sichuan, China.
- Guizhou Yibai Pharmaceutical Co. Ltd, Guiyang, 550008, Guizhou, China.
| |
Collapse
|
5
|
Xie J, Kusnadi EP, Furic L, Selth LA. Regulation of mRNA Translation by Hormone Receptors in Breast and Prostate Cancer. Cancers (Basel) 2021; 13:3254. [PMID: 34209750 PMCID: PMC8268847 DOI: 10.3390/cancers13133254] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Breast and prostate cancer are the second and third leading causes of death amongst all cancer types, respectively. Pathogenesis of these malignancies is characterised by dysregulation of sex hormone signalling pathways, mediated by the estrogen receptor-α (ER) in breast cancer and androgen receptor (AR) in prostate cancer. ER and AR are transcription factors whose aberrant function drives oncogenic transcriptional programs to promote cancer growth and progression. While ER/AR are known to stimulate cell growth and survival by modulating gene transcription, emerging findings indicate that their effects in neoplasia are also mediated by dysregulation of protein synthesis (i.e., mRNA translation). This suggests that ER/AR can coordinately perturb both transcriptional and translational programs, resulting in the establishment of proteomes that promote malignancy. In this review, we will discuss relatively understudied aspects of ER and AR activity in regulating protein synthesis as well as the potential of targeting mRNA translation in breast and prostate cancer.
Collapse
Affiliation(s)
- Jianling Xie
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia
| | - Eric P Kusnadi
- Translational Prostate Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Cancer Program, Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Luc Furic
- Translational Prostate Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Cancer Program, Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Luke A Selth
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia
- Freemasons Centre for Male Health and Wellbeing, Flinders University, Bedford Park, SA 5042, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
6
|
Chhatbar DM, Chaube UJ, Vyas VK, Bhatt HG. CoMFA, CoMSIA, Topomer CoMFA, HQSAR, molecular docking and molecular dynamics simulations study of triazine morpholino derivatives as mTOR inhibitors for the treatment of breast cancer. Comput Biol Chem 2019; 80:351-363. [DOI: 10.1016/j.compbiolchem.2019.04.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 04/26/2019] [Accepted: 04/30/2019] [Indexed: 12/20/2022]
|
7
|
Yi Z, Ma F, Liu B, Guan X, Li L, Li C, Qian H, Xu B. Everolimus in hormone receptor-positive metastatic breast cancer: PIK3CA mutation H1047R was a potential efficacy biomarker in a retrospective study. BMC Cancer 2019; 19:442. [PMID: 31088410 PMCID: PMC6515626 DOI: 10.1186/s12885-019-5668-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/01/2019] [Indexed: 02/08/2023] Open
Abstract
Background Everolimus, an inhibitor of mammalian target of rapamycin (mTOR), has been shown to increase the efficacy of endocrine therapies in hormone receptor (HR)-positive metastatic breast cancer. However, because breast cancer is a highly heterogeneous disease, the responses of different patients to everolimus may vary. Therefore, we performed this study to better select patients who will benefit most from or be resistant to everolimus. Methods Patients with HR-positive breast cancer who were treated with everolimus at the Cancer Hospital, Chinese Academy of Medical Sciences from February 2014 to March 2017 were enrolled in the present study. Mutations in ctDNA were assayed in 1021 tumor-related genes via gene panel target capture-based next-generation sequencing. Results In total, 120 patients with metastatic breast cancer who were treated with everolimus were enrolled in the present study. The median progression-free survival (PFS) of all patients was 5.1 months (95% confidence interval [CI] 3.9–6.3 months). No difference in survival was observed between patients who received endocrine drugs used in previous treatment regimens and patients who did not receive these drugs (median PFS 5.2 and 5.1 months, respectively, p > 0.05). Additionally, we did not find any difference in outcomes between patients who had primary resistance to previously used endocrine drugs and patients who had nonprimary resistance to previous treatments (p > 0.05). Multivariate analysis showed that < 3 metastatic sites, < 2 lines of previous endocrine therapy, < 2 lines of previous chemotherapy, and treatment with everolimus combined with fulvestrant were associated with improved survival (p < 0.05). Sixteen patients underwent ctDNA analysis before everolimus treatment. The frequency of PIK3CA gene mutations was 62.5%, and H1047R was the most frequently detected mutation. Patients with the PIK3CA/H1047R mutation had longer PFS than patients with wild-type or other mutant forms of PIK3CA, and the median PFS in these two groups of patients was 8.8 and 4.1 months, respectively (p < 0.05). Conclusions Our data suggest that patients who receive more lines of chemotherapy or endocrine therapy are less likely to benefit from everolimus. For everolimus combination therapy, we can even select endocrine drugs that gave rise to primary resistance in previous treatments. Additionally, the PIK3CA/H1047R mutation may be a potential biomarker of sensitivity to everolimus. Electronic supplementary material The online version of this article (10.1186/s12885-019-5668-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zongbi Yi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| | - Binliang Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Xiuwen Guan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Lixi Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Chunxiao Li
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Haili Qian
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| |
Collapse
|
8
|
mTOR Activation in Liver Tumors Is Associated with Metabolic Syndrome and Non-Alcoholic Steatohepatitis in Both Mouse Models and Humans. Cancers (Basel) 2018; 10:cancers10120465. [PMID: 30469530 PMCID: PMC6315895 DOI: 10.3390/cancers10120465] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/12/2018] [Accepted: 11/20/2018] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) can cause liver fibrosis and cirrhosis, with final progression to hepatocellular carcinoma (HCC) in some cases. Various factors have been suggested to be involved in the development of NASH. Considering the many possible contributing factors, we postulated that mechanisms of progression from NASH to HCC could differ depending on the risk factors. In the present study, we applied two mouse models of NASH⁻HCC and performed histopathological and proteome analyses of mouse liver tumors. Furthermore, to compare the mechanisms of NASH⁻HCC progression in mice and humans, we investigated HCCs in humans with a background of metabolic syndrome and NASH, as well as HCCs associated with hepatitis virus infection by immunohistochemistry. It was demonstrated that upstream regulators associated with the mammalian target of rapamycin (mTOR) pathway were altered in liver tumors of mice with metabolic syndrome characteristics (TSOD mice) using proteome analysis. Immunohistochemical analysis showed that mTOR was characteristically phosphorylated in liver tumors of TSOD mice and HCCs from metabolic syndrome cases in humans. These results indicated that the mTOR pathway is characteristically activated in liver tumors with metabolic syndrome and NASH, unlike liver tumors with other etiologies.
Collapse
|
9
|
Yi Z, Ma F. Biomarkers of Everolimus Sensitivity in Hormone Receptor-Positive Breast Cancer. J Breast Cancer 2017; 20:321-326. [PMID: 29285035 PMCID: PMC5743990 DOI: 10.4048/jbc.2017.20.4.321] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 10/08/2017] [Indexed: 01/24/2023] Open
Abstract
Activation of the mammalian target of rapamycin (mTOR) signaling pathway is an important mechanism of resistance to endocrine therapy in breast cancer. Everolimus, an mTOR inhibitor, has been shown to increase the efficacy of endocrine therapy and overcome resistance to endocrine therapies. Clinical studies have suggested that everolimus combined with endocrine therapy prolongs progression-free survival in hormone receptor-positive breast cancer patients. However, because breast cancer includes a group of highly heterogeneous tumors, patients may have different responses to everolimus. Therefore, finding biomarkers that can predict a patient's positive response or resistance to everolimus is critical. Numerous preclinical studies have shown that PIK3CA/PTEN mutations are predictive of sensitivity to everolimus; however, clinical trials have not confirmed the correlation between mutation status and clinical response. KRAS or BRAF mutations can bypass the phosphatidylinositol 3-kinase pathway; therefore, mutations in KRAS or BRAF may lead to resistance to mTOR inhibitors, and preclinical studies have shown that PIK3CA mutant cells which also contain KRAS mutations are resistant to everolimus. However, there are no clinical data in breast cancer patients to support this conclusion. Therefore, large-scale clinical studies are needed to identify biomarkers of efficacy and resistance to everolimus.
Collapse
Affiliation(s)
- Zongbi Yi
- Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
10
|
Xia R, Chen SX, Qin Q, Chen Y, Zhang WW, Zhu RR, Deng AM. Oridonin Suppresses Proliferation of Human Ovarian Cancer Cells via Blockage of mTOR Signaling. Asian Pac J Cancer Prev 2017; 17:667-71. [PMID: 26925661 DOI: 10.7314/apjcp.2016.17.2.667] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Oridonin, an ent-kaurane diterpenoid compound isolated from the traditional Chinese herb Rabdosia rubescens, has shown various pharmacological and physiological effects such as anti-tumor, anti-bacterial, and anti-inflammatory properties. However, the effect of oridonin on human ovarian cancer cell lines has not been determined. In this study, we demonstrated that oridonin inhibited ovarian cancer cell proliferation, migration and invasion in a dose-dependent manner. Furthermore, we showed oridonin inhibited tumor growth of ovarian cancer cells (SKOV3) in vivo. We then assessed mechanisms and found that oridonin specifically abrogated the phosphorylation/activation of mTOR signaling. In summary, our results indicate that oridonin is a potential inhibitor of ovarian cancer by blocking the mTOR signaling pathway.
Collapse
Affiliation(s)
- Rong Xia
- Department of Transfusion, Huashan Hospital, Fudan University, Shanghai, P. R. China E-mail :
| | | | | | | | | | | | | |
Collapse
|
11
|
Anwar SL, Wahyono A, Aryandono T, Haryono SJ. Caveolin-1 in Breast Cancer: Single Molecule Regulation of Multiple Key Signaling Pathways. Asian Pac J Cancer Prev 2016; 16:6803-12. [PMID: 26514450 DOI: 10.7314/apjcp.2015.16.16.6803] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Caveolin-1 is a 22-kD trans-membrane protein enriched in particular plasma membrane invaginations known as caveolae. Cav-1 expression is often dysregulated in human breast cancers, being commonly upregulated in cancer cells and downregulated in stromal cells. As an intracellular scaffolding protein, Cav-1, is involved in several vital biological regulations including endocytosis, transcytosis, vesicular transport, and signaling pathways. Several pathways are modulated by Cav-1 including estrogen receptor, EGFR, Her2/neu, TGFβ, and mTOR and represent as major drivers in mammary carcinogenesis. Expression and role of Cav-1 in breast carcinogenesis is highly variable depending on the stage of tumor development as well as context of the cell. However, recent data have shown that downregulation of Cav-1 expression in stromal breast tumors is associated with frequent relapse, resistance to therapy, and poor outcome. Modification of Cav-1 expression for translational cancer therapy is particularly challenging since numerous signaling pathways might be affected. This review focuses on present understanding of Cav-1 in breast carcinogenesis and its potential role as a new biomarker for predicting therapeutic response and prognosis as well as new target for therapeutic manipulation.
Collapse
Affiliation(s)
- Sumadi Lukman Anwar
- Department of Surgery, Faculty of Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia E-mail :
| | | | | | | |
Collapse
|
12
|
Behrouz H, Esfandyari-Manesh M, Khoeeniha MK, Amini M, Shiri Varnamkhasti B, Atyabi F, Dinarvand R. Enhanced Cytotoxicity to Cancer Cells by Codelivery and Controlled Release of Paclitaxel-loaded Sirolimus-conjugated Albumin Nanoparticles. Chem Biol Drug Des 2016; 88:230-40. [DOI: 10.1111/cbdd.12750] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 12/19/2015] [Accepted: 02/08/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Hossein Behrouz
- Department of Pharmaceutics; Faculty of Pharmacy; Tehran University of Medical Sciences; P.O. Box 14155-6451 Tehran Iran
| | - Mehdi Esfandyari-Manesh
- Nanotechnology Research Center; Faculty of Pharmacy; Tehran University of Medical Sciences; P.O. Box 14155-6451 Tehran Iran
| | - Mohammad Kazem Khoeeniha
- Department of Pharmaceutics; Faculty of Pharmacy; Tehran University of Medical Sciences; P.O. Box 14155-6451 Tehran Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry; Faculty of Pharmacy; Tehran University of Medical Sciences; P.O. Box 14155-6451 Tehran Iran
| | - Behrang Shiri Varnamkhasti
- Nanotechnology Research Center; Faculty of Pharmacy; Tehran University of Medical Sciences; P.O. Box 14155-6451 Tehran Iran
| | - Fatemeh Atyabi
- Department of Pharmaceutics; Faculty of Pharmacy; Tehran University of Medical Sciences; P.O. Box 14155-6451 Tehran Iran
- Nanotechnology Research Center; Faculty of Pharmacy; Tehran University of Medical Sciences; P.O. Box 14155-6451 Tehran Iran
| | - Rassoul Dinarvand
- Department of Pharmaceutics; Faculty of Pharmacy; Tehran University of Medical Sciences; P.O. Box 14155-6451 Tehran Iran
- Nanotechnology Research Center; Faculty of Pharmacy; Tehran University of Medical Sciences; P.O. Box 14155-6451 Tehran Iran
| |
Collapse
|
13
|
Deng K, Yu L, Zheng X, Zhang K, Wang W, Dong P, Zhang J, Ren M. Target of Rapamycin Is a Key Player for Auxin Signaling Transduction in Arabidopsis. FRONTIERS IN PLANT SCIENCE 2016; 7:291. [PMID: 27014314 PMCID: PMC4786968 DOI: 10.3389/fpls.2016.00291] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 02/23/2016] [Indexed: 05/08/2023]
Abstract
Target of rapamycin (TOR), a master sensor for growth factors and nutrition availability in eukaryotic species, is a specific target protein of rapamycin. Rapamycin inhibits TOR kinase activity viaFK506 binding protein 12 kDa (FKBP12) in all examined heterotrophic eukaryotic organisms. In Arabidopsis, several independent studies have shown that AtFKBP12 is non-functional under aerobic condition, but one study suggests that AtFKBP12 is functional during anaerobic growth. However, the functions of AtFKBP12 have never been examined in parallel under aerobic and anaerobic growth conditions so far. To this end, we cloned the FKBP12 gene of humans, yeast, and Arabidopsis, respectively. Transgenic plants were generated, and pharmacological examinations were performed in parallel with Arabidopsis under aerobic and anaerobic conditions. ScFKBP12 conferred plants with the strongest sensitivity to rapamycin, followed by HsFKBP12, whereas AtFKBP12 failed to generate rapamycin sensitivity under aerobic condition. Upon submergence, yeast and human FKBP12 can significantly block cotyledon greening while Arabidopsis FKBP12 only retards plant growth in the presence of rapamycin, suggesting that hypoxia stress could partially restore the functions of AtFKBP12 to bridge the interaction between rapamycin and TOR. To further determine if communication between TOR and auxin signaling exists in plants, yeast FKBP12 was introduced into DR5::GUS homozygous plants. The transgenic plants DR5/BP12 were then treated with rapamycin or KU63794 (a new inhibitor of TOR). GUS staining showed that the auxin content of root tips decreased compared to the control. DR5/BP12 plants lost sensitivity to auxin after treatment with rapamycin. Auxin-defective phenotypes, including short primary roots, fewer lateral roots, and loss of gravitropism, occurred in DR5/BP12 plants when seedlings were treated with rapamycin+KU63794. This indicated that the combination of rapamycin and KU63794 can significantly inhibit TOR and auxin signaling in DR5/BP12 plants. These studies demonstrate that TOR is essential for auxin signaling transduction in Arabidopsis.
Collapse
Affiliation(s)
- Kexuan Deng
- School of Life Sciences, Chongqing UniversityChongqing, China
| | - Lihua Yu
- School of Life Sciences, Chongqing UniversityChongqing, China
| | - Xianzhe Zheng
- School of Life Sciences, Chongqing UniversityChongqing, China
| | - Kang Zhang
- School of Life Sciences, Chongqing UniversityChongqing, China
| | - Wanjing Wang
- College of Agronomy and Biotechnology, Southwest UniversityChongqing, China
| | - Pan Dong
- School of Life Sciences, Chongqing UniversityChongqing, China
| | - Jiankui Zhang
- College of Agronomy and Biotechnology, Southwest UniversityChongqing, China
| | - Maozhi Ren
- School of Life Sciences, Chongqing UniversityChongqing, China
- *Correspondence: Maozhi Ren
| |
Collapse
|
14
|
Liu NB, Zhang JH, Liu YF, Li J, Zhang ZZ, Li JW, Liu WY, Huang C, Shen T, Gu CW, Gao DY, Wu X, Wu X. High DEPTOR expression correlates with poor prognosis in patients with esophageal squamous cell carcinoma. Onco Targets Ther 2015; 8:3449-55. [PMID: 26640385 PMCID: PMC4657798 DOI: 10.2147/ott.s92862] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE The disheveled, Egl-10, and pleckstrin (DEP) domain containing mammalian target of rapamycin (mTOR)-interacting protein (DEPTOR) is a binding protein containing mTOR complex 1 (mTORC1), mTOR complex 2 (mTORC2), and an endogenous mTOR inhibitor. DEPTOR shows abnormal expressions in numerous types of solid tumors. However, how DEP-TOR is expressed in esophageal squamous cell carcinoma (ESCC) remains elusive. METHODS The expression of DEPTOR in 220 cases of ESCC and non-cancerous adjacent tissues was detected by immunohistochemistry. DEPTOR levels in ESCC and paired normal tissue were quantified using reverse transcription-polymerase chain reaction and Western blot analysis to verify the immunohistochemical results. The relationship between DEPTOR expression and the clinicopathological features of ESCC was analyzed based on the results of immunohistochemistry. Finally, we analyzed the relationship between DEPTOR expression and the prognosis of patients with ESCC. RESULTS Immunohistochemical staining showed that the expression rate of DEPTOR in ESCC tissues was significantly increased. DEPTOR mRNA and protein expression was significantly higher in ESCC tissues than in normal adjacent esophageal squamous tissues. High DEPTOR expression was significantly correlated with regional lymph node status in the TNM stage of patients with ESCC. Kaplan-Meier survival curves showed that the rate of overall survival was significantly lower in patients with high DEPTOR expression than in those with low DEPTOR expression. Additionally, high DEPTOR expression was an independent prognostic predictor for ESCC patients. CONCLUSION High DEPTOR expression is an independent prognostic biomarker indicating a worse prognosis for patients with ESCC.
Collapse
Affiliation(s)
- Nan-Bo Liu
- Department of Thoracic Surgery, Southern Medical University, Guangzhou, People's Republic of China
| | - Jun-Hua Zhang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Yu-Fan Liu
- Department of Thoracic Surgery, Southern Medical University, Guangzhou, People's Republic of China
| | - Jun Li
- Department of Thoracic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, People's Republic of China
| | - Zhen-Zhong Zhang
- Department of Thoracic Surgery, Southern Medical University, Guangzhou, People's Republic of China
| | - Ji-Wei Li
- Department of Thoracic Surgery, Southern Medical University, Guangzhou, People's Republic of China
| | - Wen-Yue Liu
- Department of Thoracic Surgery, Southern Medical University, Guangzhou, People's Republic of China
| | - Chen Huang
- Department of Thoracic Surgery, Southern Medical University, Guangzhou, People's Republic of China ; Department of Thoracic Surgery, Fujian Provincial Hospital, Fuzhou, People's Republic of China
| | - Tao Shen
- Department of Thoracic Surgery, Jiangmen Central Hospital, Jiangmen, People's Republic of China
| | - Cheng-Wei Gu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, People's Republic of China
| | - Dong-Yun Gao
- Department of Oncology, Dongtai People's Hospital, Dongtai, People's Republic of China
| | - Xia Wu
- Department of Breast Cancer, Affiliated Hospital, Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Xu Wu
- Department of Thoracic Surgery, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
15
|
Generali D, Berruti A, Cappelletti MR, Zanotti L, Brugnoli G, Forti M, Bedussi F, Vailati ME, Milani M, Strina C, Ardine M, Aguggini S, Allevi G, Ferrero G, Bertoni R, Bottini A, Harris AL, Fox SB. Effect of Primary Letrozole Treatment on Tumor Expression of mTOR and HIF-1α and Relation to Clinical Response. J Natl Cancer Inst Monogr 2015; 2015:64-6. [PMID: 26063890 DOI: 10.1093/jncimonographs/lgv018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2024] Open
Abstract
INTRODUCTION Recently the combination of the mammalian target of rapamycin (mTOR) inhibitor everolimus and the aromatase inhibitor exemestane has been shown to double the progression-free survival rate in advanced breast cancer. However, the effect of the interrelated pathways of hypoxia-inducible factor-1α (HIF-1α) and mTOR signaling, both of which are associated with a more aggressive breast cancer phenotype and endocrine resistance, on response in the neoadjuvant setting is unknown. We, therefore, have investigated the influence of these pathways with the aim of better defining those patients most likely to benefit from an endocrine-based therapy associated with/without mTOR inhibitors. PATIENTS AND METHODS A total of 107 women with T2-4 N0-1 and estrogen receptor-positive breast cancer were randomly assigned to 6 months of primary letrozole (2.5 mg/daily) (LET) or LET plus oral "metronomic" cyclophosphamide (50mg/daily) (LET-CYC). Phospo-mTOR and HIF-1α were evaluated in tumor specimens collected before and after treatment using a tissue microarray format. RESULTS LET-based therapy induced a downregulation of phospho-mTOR and HIF-1α expression (P = .0001 and P < .004, respectively). The reduction of HIF-1α expression observed was positively correlated with phospho-mTOR reduction (P < .03); however, no treatment interaction between the two proteins was detected. HIF-1α expression was significantly modulated by the treatment (P < .004) with a reduction both in the LET arm (45%, n = 36/80) (P = .05) and LET-CYC arm (55%, n = 44/80) (P = .04). HIF-1α reduction showed a relationship with clinical response confined in LET arm only (P < .03). CONCLUSIONS In this neoadjuvant population, LET was able to modulate the phospho-mTOR and HIF-1α pathways and may define a subpopulation of nonresponders who may be most likely to benefit from mTOR inhibitors.
Collapse
Affiliation(s)
- Daniele Generali
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF).
| | - Alfredo Berruti
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Maria Rosa Cappelletti
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Laura Zanotti
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Giulia Brugnoli
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Michela Forti
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Francesca Bedussi
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Maria Elena Vailati
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Manuela Milani
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Carla Strina
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Mara Ardine
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Sergio Aguggini
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Giovanni Allevi
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Giuseppina Ferrero
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Ramona Bertoni
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Alberto Bottini
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Adrian L Harris
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| | - Stephen B Fox
- U.O. Multidisciplinare di Patologia Mammaria/ US Terapia Molecolare (DG, MRC, LZ, GB, MF, FB, MEV, MM, CS, MA, SA, GA, ABo), and Anatomia Patologica (GF, RB), Azienda Istituti Ospitalieri di Cremona, Cremona, Italy; Oncologia Medica, Università di Brescia, Spedali Civili di Brescia, Brescia, Italy (ABe, GB); Weatherall Molecular Oncology Laboratories, Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK (ALH); Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Australia (SBF)
| |
Collapse
|
16
|
Correlation of perfusion MRI and 18F-FDG PET imaging biomarkers for monitoring regorafenib therapy in experimental colon carcinomas with immunohistochemical validation. PLoS One 2015; 10:e0115543. [PMID: 25668193 PMCID: PMC4323201 DOI: 10.1371/journal.pone.0115543] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 11/25/2014] [Indexed: 12/15/2022] Open
Abstract
Objectives To investigate a multimodal, multiparametric perfusion MRI / 18F-fluoro-deoxyglucose-(18F-FDG)-PET imaging protocol for monitoring regorafenib therapy effects on experimental colorectal adenocarcinomas in rats with immunohistochemical validation. Materials and Methods Human colorectal adenocarcinoma xenografts (HT-29) were implanted subcutaneously in n = 17 (n = 10 therapy group; n = 7 control group) female athymic nude rats (Hsd:RH-Foxn1rnu). Animals were imaged at baseline and after a one-week daily treatment protocol with regorafenib (10 mg/kg bodyweight) using a multimodal, multiparametric perfusion MRI/18F-FDG-PET imaging protocol. In perfusion MRI, quantitative parameters of plasma flow (PF, mL/100 mL/min), plasma volume (PV, %) and endothelial permeability-surface area product (PS, mL/100 mL/min) were calculated. In 18F-FDG-PET, tumor-to-background-ratio (TTB) was calculated. Perfusion MRI parameters were correlated with TTB and immunohistochemical assessments of tumor microvascular density (CD-31) and cell proliferation (Ki-67). Results Regorafenib significantly (p<0.01) suppressed PF (81.1±7.5 to 50.6±16.0 mL/100mL/min), PV (12.1±3.6 to 7.5±1.6%) and PS (13.6±3.2 to 7.9±2.3 mL/100mL/min) as well as TTB (3.4±0.6 to 1.9±1.1) between baseline and day 7. Immunohistochemistry revealed significantly (p<0.03) lower tumor microvascular density (CD-31, 7.0±2.4 vs. 16.1±5.9) and tumor cell proliferation (Ki-67, 434.0 ± 62.9 vs. 663.0 ± 98.3) in the therapy group. Perfusion MRI parameters ΔPF, ΔPV and ΔPS showed strong and significant (r = 0.67-0.78; p<0.01) correlations to the PET parameter ΔTTB and significant correlations (r = 0.57-0.67; p<0.03) to immunohistochemical Ki-67 as well as to CD-31-stainings (r = 0.49-0.55; p<0.05). Conclusions A multimodal, multiparametric perfusion MRI/PET imaging protocol allowed for non-invasive monitoring of regorafenib therapy effects on experimental colorectal adenocarcinomas in vivo with significant correlations between perfusion MRI parameters and 18F-FDG-PET validated by immunohistochemistry.
Collapse
|
17
|
Selvarajah J, Elia A, Carroll VA, Moumen A. DNA damage-induced S and G2/M cell cycle arrest requires mTORC2-dependent regulation of Chk1. Oncotarget 2015; 6:427-40. [PMID: 25460505 PMCID: PMC4381605 DOI: 10.18632/oncotarget.2813] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 11/14/2014] [Indexed: 12/25/2022] Open
Abstract
mTOR signalling is commonly dysregulated in cancer. Concordantly, mTOR inhibitors have demonstrated efficacy in a subset of tumors and are in clinical trials as combination therapies. Although mTOR is associated with promoting cell survival after DNA damage, the exact mechanisms are not well understood. Moreover, since mTOR exists as two complexes, mTORC1 and mTORC2, the role of mTORC2 in cancer and in the DNA damage response is less well explored. Here, we report that mTOR protein levels and kinase activity are transiently increased by DNA damage in an ATM and ATR-dependent manner. We show that inactivation of mTOR with siRNA or pharmacological inhibition of mTORC1/2 kinase prevents etoposide-induced S and G2/M cell cycle arrest. Further results show that Chk1, a key regulator of the cell cycle arrest, is important for this since ablation of mTOR prevents DNA damage-induced Chk1 phosphorylation and decreases Chk1 protein production. Furthermore, mTORC2 was essential and mTORC1 dispensable, for this role. Importantly, we show that mTORC1/2 inhibition sensitizes breast cancer cells to chemotherapy. Taken together, these results suggest that breast cancer cells may rely on mTORC2-Chk1 pathway for survival and provide evidence that mTOR kinase inhibitors may overcome resistance to DNA-damage based therapies in breast cancer.
Collapse
Affiliation(s)
- Jogitha Selvarajah
- Cardiovascular and Cell Sciences Research Institute, St George's University of London, Cranmer Terrace, UK
| | - Androulla Elia
- Cardiovascular and Cell Sciences Research Institute, St George's University of London, Cranmer Terrace, UK
| | - Veronica A. Carroll
- Cardiovascular and Cell Sciences Research Institute, St George's University of London, Cranmer Terrace, UK
| | - Abdeladim Moumen
- Division of Medical Biotechnology, MAscIR Institution, Rabat, Morocco
| |
Collapse
|
18
|
Ou O, Huppi K, Chakka S, Gehlhaus K, Dubois W, Patel J, Chen J, Mackiewicz M, Jones TL, Pitt JJ, Martin SE, Goldsmith P, Simmons JK, Mock BA, Caplen NJ. Loss-of-function RNAi screens in breast cancer cells identify AURKB, PLK1, PIK3R1, MAPK12, PRKD2, and PTK6 as sensitizing targets of rapamycin activity. Cancer Lett 2014; 354:336-47. [PMID: 25193464 PMCID: PMC4240001 DOI: 10.1016/j.canlet.2014.08.043] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 08/15/2014] [Accepted: 08/22/2014] [Indexed: 02/05/2023]
Abstract
The use of molecularly targeted drugs as single agents has shown limited utility in many tumor types, largely due to the complex and redundant nature of oncogenic signaling networks. Targeting of the PI3K/AKT/mTOR pathway through inhibition of mTOR in combination with aromatase inhibitors has seen success in particular sub-types of breast cancer and there is a need to identify additional synergistic combinations to maximize the clinical potential of mTOR inhibitors. We have used loss-of-function RNAi screens of the mTOR inhibitor rapamycin to identify sensitizers of mTOR inhibition. RNAi screens conducted in combination with rapamycin in multiple breast cancer cell lines identified six genes, AURKB, PLK1, PIK3R1, MAPK12, PRKD2, and PTK6 that when silenced, each enhanced the sensitivity of multiple breast cancer lines to rapamycin. Using selective pharmacological agents we confirmed that inhibition of AURKB or PLK1 synergizes with rapamycin. Compound-associated gene expression data suggested histone deacetylation (HDAC) inhibition as a strategy for reducing the expression of several of the rapamycin-sensitizing genes, and we tested and validated this using the HDAC inhibitor entinostat in vitro and in vivo. Our findings indicate new approaches for enhancing the efficacy of rapamycin including the use of combining its application with HDAC inhibition.
Collapse
Affiliation(s)
- Oliver Ou
- Genetics Branch, National Institutes of Health, Bethesda, MD 20892, USA
| | - Konrad Huppi
- Genetics Branch, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sirisha Chakka
- Genetics Branch, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kristen Gehlhaus
- Genetics Branch, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wendy Dubois
- Laboratory of Cancer Biology and Genetics, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jyoti Patel
- Laboratory of Cancer Biology and Genetics, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jinqiu Chen
- Office of Science and Technology Partnerships, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mark Mackiewicz
- Genetics Branch, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tamara L Jones
- Genetics Branch, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jason J Pitt
- Genetics Branch, National Institutes of Health, Bethesda, MD 20892, USA
| | - Scott E Martin
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20850, USA
| | - Paul Goldsmith
- Office of Science and Technology Partnerships, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - John K Simmons
- Laboratory of Cancer Biology and Genetics, National Institutes of Health, Bethesda, MD 20892, USA
| | - Beverly A Mock
- Laboratory of Cancer Biology and Genetics, National Institutes of Health, Bethesda, MD 20892, USA
| | - Natasha J Caplen
- Genetics Branch, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
19
|
Alex JM, Singh S, Kumar R. 1-Acetyl-3,5-diaryl-4,5-dihydro(1H)pyrazoles: Exhibiting Anticancer Activity through Intracellular ROS Scavenging and the Mitochondria-Dependent Death Pathway. Arch Pharm (Weinheim) 2014; 347:717-27. [DOI: 10.1002/ardp.201400199] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Revised: 06/19/2014] [Accepted: 06/24/2014] [Indexed: 11/07/2022]
Affiliation(s)
- Jimi M. Alex
- Laboratory for Drug Design and Synthesis; Centre for Chemical and Pharmaceutical Sciences; School of Basic and Applied Sciences; Central University of Punjab; Bathinda India
| | - Sandeep Singh
- Centre for Genetic Diseases and Molecular Medicine; Central University of Punjab; Bathinda India
| | - Raj Kumar
- Laboratory for Drug Design and Synthesis; Centre for Chemical and Pharmaceutical Sciences; School of Basic and Applied Sciences; Central University of Punjab; Bathinda India
| |
Collapse
|
20
|
Ghanbarzadeh S, Khorrami A, Mohamed Khosroshahi L, Arami S. Fusogenic pH sensitive liposomal formulation for rapamycin: improvement of antiproliferative effect. PHARMACEUTICAL BIOLOGY 2014; 52:848-854. [PMID: 24920230 DOI: 10.3109/13880209.2013.871640] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
CONTEXT Liposomes are increasingly employed to deliver chemotherapeutic agents, antisense oligonucleotides, and genes to various therapeutic targets. OBJECTIVE The present investigation evaluates the ability of fusogenic pH-sensitive liposomes of rapamycin in increasing its antiproliferative effect on human breast adenocarcinoma (MCF-7) cell line. MATERIALS AND METHODS Cholesterol (Chol) and dipalmitoylphosphatidylcholine (DPPC) (DPPC:Chol, 7:3) were used to prepare conventional rapamycin liposomes by a modified ethanol injection method. Dioleoylphosphatidylethanolamine (DOPE) was used to produce fusogenic and pH-sensitive properties in liposomes simultaneously (DPPC:Chol:DOPE, 7:3:4.2). The prepared liposomes were characterized by their size, zeta potential, encapsulation efficiency percent (EE%), and chemical stability during 6 months. The antiproliferative effects of both types of rapamycin liposomes (10, 25, and 50 nmol/L) with optimized formulations were assessed on MCF-7 cells, as cancerous cells, and human umbilical vein endothelial cells (HUVEC), as healthy cells, employing the diphenyltetrazolium bromide (MTT) assay for 72 h. RESULTS AND DISCUSSION The particle size, zeta potential, and EE% of the liposomes were 165 ± 12.3 and 178 ± 15.4 nm, -39.6 ± 1.3, and -41.2 ± 2.1 mV as well as 76.9 ± 2.6 and 76.9 ± 2.6% in conventional and fusogenic pH-sensitive liposomes, respectively. Physicochemical stability results indicated that both liposome types were relatively stable at 4 °C than 25 °C. In vitro antiproliferative evaluation showed that fusogenic pH-sensitive liposomes had better antiproliferative effects on MCF-7 cells compared to the conventional liposomes. Conversely, fusogenic pH-sensitive liposomes had less cytotoxicity on HUVEC cell line.
Collapse
Affiliation(s)
- Saeed Ghanbarzadeh
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences , Tabriz , Iran
| | | | | | | |
Collapse
|
21
|
Zhao N, Li X, He X, Qiu Y, Zhu L, Qi F. Interleukin-15 gene therapy and the mammalian target of rapamycin inhibitor everolimus inhibit the growth of metastatic breast cancer. J Gene Med 2014; 15:366-74. [PMID: 24038990 DOI: 10.1002/jgm.2739] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 08/10/2013] [Accepted: 08/27/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Novel methods to control and treat metastatic breast cancer are needed. Interleukin (IL)-15 is a promising cytokine for cancer immunotherapy, and everolimus is an orally administered mammalian target of rapamycin (mTOR) inhibitor, which is already approved for cancer treatment. In the present study, we investigated the efficacy of IL-15 gene therapy and explored the possibility of combining IL-15 therapy with everolimus to treat metastatic breast cancer. METHODS A plasmid encoding IL-15 and everolimus were given to mice inoculated with 4 T1 mouse breast cancer cells. Tumor size and metastasis were monitored to assess the effect of different treatment regimens. Immunohistochemistry was used to detect CD4⁺, CD8⁺ and NKG2D⁺ cells and also the expression of Ki-67 in tumor tissue; these analyses helped establish the immunization status and tumor proliferation rate of different treatment groups. Terminal deoxynucleotidyl transferase dUTP nick end labeling assays were performed to assess cellular apoptosis in tumor tissues. RESULTS Both IL-15 and everolimus significantly decreased tumor size. IL-15 gene therapy increased the proportion of CD4⁺ T and natural killer (NK) cells but had no effect on CD8⁺ T cells. By contrast, everolimus decreased the number of CD8⁺ T cells but had no effect on CD4⁺ T and NK cells compared to the control group. Both IL-15 and everolimus decreased expression of Ki-67 and increased rates of apoptosis. Although effective on their own, no synergistic effect was observed with a combined treatment of everolimus and IL-15 gene therapy. CONCLUSIONS IL-15 gene therapy was potentially useful for the treatment of metastatic breast cancer. The possibility of combining immunotherapy with everolimus requires further study.
Collapse
Affiliation(s)
- Na Zhao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | | | | | | | | | | |
Collapse
|
22
|
Ferrario C, Batist G. Advances in the approach to novel drug clinical development for breast cancer. Expert Opin Drug Discov 2014; 9:647-68. [PMID: 24758225 DOI: 10.1517/17460441.2014.911282] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION In the post-genomic era clinical development of new agents to treat breast cancer (BC) can be a real challenge. Different from chemotherapy agents, with a broad but not specific spectrum of activity, novel drugs are being developed as 'targeted' agents, potentially benefiting a subgroup of patients. In BC, different clinically identifiable subtypes are now separately addressed in specific clinical trials. AREAS COVERED In this review, the authors discuss the clinical development of targeted drugs that have become part of the current treatment of BC. They also highlight the challenges that in other cases determined the failure of promising compounds. Furthermore, the article reports on how combinations of targeted agents have emerged as valid strategies to overcome acquired resistance. It also provides discussion of how 'old' therapies can be retargeted to certain patient populations or 'reinvented' as safer and more effective with the creation of drug conjugates. They also discuss how novel clinical trial designs are emerging to accelerate the successful matching of targeted drugs to the right patient population. EXPERT OPINION It is important not to forget that the development of BC therapeutics is a 'moving target', as its biology evolves in time under the pressure of ongoing treatments. There are currently a finite number of resources available for the development of new therapeutics, which means that resources need to be carefully allocated. There is also a need to prioritize clinical trials that can reduce the number of patients who are candidates for expensive treatments.
Collapse
Affiliation(s)
- Cristiano Ferrario
- McGill University, Jewish General Hospital, Segal Cancer Centre, Department of Oncology , 3755 Cote Ste Catherine Rd. W, Montreal, Quebec H3T1E2 , Canada
| | | |
Collapse
|
23
|
Sahin K, Orhan C, Tuzcu M, Sahin N, Ali S, Bahcecioglu IH, Guler O, Ozercan I, Ilhan N, Kucuk O. Orally Administered Lycopene Attenuates Diethylnitrosamine-Induced Hepatocarcinogenesis in Rats by Modulating Nrf-2/HO-1 and Akt/mTOR Pathways. Nutr Cancer 2014; 66:590-8. [DOI: 10.1080/01635581.2014.894092] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
24
|
Chang FY, Brady SF. Characterization of an environmental DNA-derived gene cluster that encodes the bisindolylmaleimide methylarcyriarubin. Chembiochem 2014; 15:815-21. [PMID: 24648189 DOI: 10.1002/cbic.201300756] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Indexed: 01/11/2023]
Abstract
Bisindolylmaleimides represent a naturally occurring class of metabolites that are of interest because of their protein kinase inhibition activity. From a metagenomic library constructed with soil DNA, we identified the four gene mar cluster, a bisindolylmaleimide gene cluster that encodes for methylarcyriarubin (1) production. Heterologous expression of the mar gene cluster in E. coli revealed that the Rieske dioxygenase MarC facilitates the oxidative decarboxylation of a chromopyrrolic acid (CPA) intermediate to yield the bisindolylmaleimide core. The characterization of the mar cluster defines a new role for CPA in the biosynthesis of structurally diverse bacterial tryptophan dimers.
Collapse
Affiliation(s)
- Fang-Yuan Chang
- Laboratory of Genetically Encoded Small Molecules, Howard Hughes Medical Institute, The Rockefeller University, 1230 York Avenue, New York, NY 10065 (USA)
| | | |
Collapse
|
25
|
Kümler I, Tuxen MK, Nielsen DL. A systematic review of dual targeting in HER2-positive breast cancer. Cancer Treat Rev 2014; 40:259-70. [DOI: 10.1016/j.ctrv.2013.09.002] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 08/26/2013] [Accepted: 09/02/2013] [Indexed: 11/30/2022]
|
26
|
Jordan NJ, Dutkowski CM, Barrow D, Mottram HJ, Hutcheson IR, Nicholson RI, Guichard SM, Gee JMW. Impact of dual mTORC1/2 mTOR kinase inhibitor AZD8055 on acquired endocrine resistance in breast cancer in vitro. Breast Cancer Res 2014; 16:R12. [PMID: 24457069 PMCID: PMC3978713 DOI: 10.1186/bcr3604] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 01/15/2014] [Indexed: 12/18/2022] Open
Abstract
Introduction Upregulation of PI3K/Akt/mTOR signalling in endocrine-resistant breast cancer (BC) has identified mTOR as an attractive target alongside anti-hormones to control resistance. RAD001 (everolimus/Afinitor®), an allosteric mTOR inhibitor, is proving valuable in this setting; however, some patients are inherently refractory or relapse during treatment requiring alternative strategies. Here we evaluate the potential for novel dual mTORC1/2 mTOR kinase inhibitors, exemplified by AZD8055, by comparison with RAD001 in ER + endocrine resistant BC cells. Methods In vitro models of tamoxifen (TamR) or oestrogen deprivation resistance (MCF7-X) were treated with RAD001 or AZD8055 alone or combined with anti-hormone fulvestrant. Endpoints included growth, cell proliferation (Ki67), viability and migration, with PI3K/AKT/mTOR signalling impact monitored by Western blotting. Potential ER cross-talk was investigated by immunocytochemistry and RT-PCR. Results RAD001 was a poor growth inhibitor of MCF7-derived TamR and MCF7-X cells (IC50 ≥1 μM), rapidly inhibiting mTORC1 but not mTORC2/AKT signalling. In contrast AZD8055, which rapidly inhibited both mTORC1 and mTORC2/AKT activity, was a highly effective (P <0.001) growth inhibitor of TamR (IC50 18 nM) and MCF7-X (IC50 24 nM), and of a further T47D-derived tamoxifen resistant model T47D-tamR (IC50 19 nM). AZD8055 significantly (P <0.05) inhibited resistant cell proliferation, increased cell death and reduced migration. Furthermore, dual treatment of TamR or MCF7-X cells with AZD8055 plus fulvestrant provided superior control of resistant growth versus either agent alone (P <0.05). Co-treating with AZD8055 alongside tamoxifen (P <0.01) or oestrogen deprivation (P <0.05) also effectively inhibited endocrine responsive MCF-7 cells. Although AZD8055 inhibited oestrogen receptor (ER) ser167 phosphorylation in TamR and MCF7-X, it had no effect on ER ser118 activity or expression of several ER-regulated genes, suggesting the mTOR kinase inhibitor impact was largely ER-independent. The capacity of AZD8055 for ER-independent activity was further evidenced by growth inhibition (IC5018 and 20 nM) of two acquired fulvestrant resistant models lacking ER. Conclusions This is the first report demonstrating dual mTORC1/2 mTOR kinase inhibitors have potential to control acquired endocrine resistant BC, even under conditions where everolimus fails. Such inhibitors may prove of particular benefit when used alongside anti-hormonal treatment as second-line therapy in endocrine resistant disease, and also potentially alongside anti-hormones during the earlier endocrine responsive phase to hinder development of resistance.
Collapse
|
27
|
Abstract
BACKGROUND Breast cancer cells can develop resistance to standard hormonal treatment and chemotherapy with the activation of the mTOR pathway; this is supported by results of preclinical and clinical studies. In clinical trials, the addition of everolimus to hormonal treatment or anti-HER2 treatment improved the outcomes of breast cancer patients. The aim of this review is to discuss the efficacy and safety data of everolimus in all categories of breast cancer in recent published studies. SCOPE Everolimus showed positive results in clinical studies. A literature search was made from PubMed, ASCO and San Antonio Breast Cancer Symposium Meeting abstracts by using the following search key words: 'everolimus', 'RAD001', 'mTOR inhibitor', 'breast cancer' 'endocrine therapy resistance' and 'HER-2 targeted therapies'. The last search was on June 10, 2013. The most important limitation of our review is that most of the data on everolimus rely on phase I and II trials. FINDINGS Preclinical studies showed that mTOR activation can be the responsible mechanism in all subgroups of breast cancer. Results of both the TAMRAD and BOLERO-2 studies have showed that mTOR inhibition in combination with endocrine therapy can be a new treatment strategy for MBC patients who are resistant to aromatase inhibitors. In the BOLERO-2 study, time to deterioration in health-related quality of life was also significantly higher in the everolimus and exemestane arm compared to the exemestane plus placebo arm. The recently completed BOLERO-3 study showed that mTOR inhibition in combination with trastuzumab plus vinorelbine treatment significantly improved PFS compared to trastuzumab plus vinorelbine alone in trastuzumab-resistant MBC patients. CONCLUSION Recent trials have shown that everolimus has produced promising anti-tumor activity in combination with trastuzumab in HER2-positive metastatic breast cancer and in combination with exemestane in patients with hormone-receptor-positive metastatic breast cancer who had recurrence or progression while receiving a nonsteroidal aromatase inhibitor. Results of ongoing studies with everolimus show evidence that using everolimus in earlier stages of the disease, namely in the adjuvant and neoadjuvant settings, could be benefical.
Collapse
Affiliation(s)
- Mehmet A N Sendur
- Ankara Numune Education and Research Hospital, Department of Medical Oncology , Ankara , Turkey
| | | | | | | |
Collapse
|
28
|
Romano G. The role of the dysfunctional akt-related pathway in cancer: establishment and maintenance of a malignant cell phenotype, resistance to therapy, and future strategies for drug development. SCIENTIFICA 2013; 2013:317186. [PMID: 24381788 PMCID: PMC3870877 DOI: 10.1155/2013/317186] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 11/14/2013] [Indexed: 06/01/2023]
Abstract
Akt serine/threonine kinases, or PKB, are key players in the regulation of a wide variety of cellular activities, such as growth, proliferation, protection from apoptotic injuries, control of DNA damage responses and genome stability, metabolism, migration, and angiogenesis. The Akt-related pathway responds to the stimulation mediated by growth factors, cytokines, hormones, and several nutrients. Akt is present in three isoforms: Akt1, Akt2, and Akt3, which may be alternatively named PKB α , PKB β , and PKB γ , respectively. The Akt isoforms are encoded on three diverse chromosomes and their biological functions are predominantly distinct. Deregulations in the Akt-related pathway were observed in many human maladies, including cancer, cardiopathies, neurological diseases, and type-2 diabetes. This review discusses the significance of the abnormal activities of the Akt axis in promoting and sustaining malignancies, along with the development of tumor cell populations that exhibit enhanced resistance to chemo- and/or radiotherapy. This occurrence may be responsible for the relapse of the disease, which is unfortunately very often related to fatal consequences in patients.
Collapse
Affiliation(s)
- Gaetano Romano
- Department of Biology, College of Science and Technology, Temple University, Bio Life Science Building, Suite 456, 1900 N. 12th Street, Philadelphia, PA 19122, USA
| |
Collapse
|
29
|
Cyran CC, Kazmierczak PM, Hirner H, Moser M, Ingrisch M, Havla L, Michels A, Eschbach R, Schwarz B, Reiser MF, Bruns CJ, Nikolaou K. Regorafenib effects on human colon carcinoma xenografts monitored by dynamic contrast-enhanced computed tomography with immunohistochemical validation. PLoS One 2013; 8:e76009. [PMID: 24098755 PMCID: PMC3786893 DOI: 10.1371/journal.pone.0076009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Accepted: 08/19/2013] [Indexed: 11/20/2022] Open
Abstract
Objective To investigate dynamic contrast-enhanced computed tomography for monitoring the effects of regorafenib on experimental colon carcinomas in rats by quantitative assessments of tumor microcirculation parameters with immunohistochemical validation. Materials and Methods Colon carcinoma xenografts (HT-29) implanted subcutaneously in female athymic rats (n = 15) were imaged at baseline and after a one-week treatment with regorafenib by dynamic contrast-enhanced computed tomography (128-slice dual-source computed tomography). The therapy group (n = 7) received regorafenib daily (10 mg/kg bodyweight). Quantitative parameters of tumor microcirculation (plasma flow, mL/100 mL/min), endothelial permeability (PS, mL/100 mL/min), and tumor vascularity (plasma volume, %) were calculated using a 2-compartment uptake model. Dynamic contrast-enhanced computed tomography parameters were validated with immunohistochemical assessments of tumor microvascular density (CD-31), tumor cell apoptosis (TUNEL), and proliferation (Ki-67). Results Regorafenib suppressed tumor vascularity (15.7±5.3 to 5.5±3.5%; p<0.05) and tumor perfusion (12.8±2.3 to 8.8±2.9 mL/100 mL/min; p = 0.063). Significantly lower microvascular density was observed in the therapy group (CD-31; 48±10 vs. 113±25, p<0.05). In regorafenib-treated tumors, significantly more apoptotic cells (TUNEL; 11844±2927 vs. 5097±3463, p<0.05) were observed. Dynamic contrast-enhanced computed tomography tumor perfusion and tumor vascularity correlated significantly (p<0.05) with microvascular density (CD-31; r = 0.84 and 0.66) and inversely with apoptosis (TUNEL; r = −0.66 and −0.71). Conclusions Regorafenib significantly suppressed tumor vascularity (plasma volume) quantified by dynamic contrast-enhanced computed tomography in experimental colon carcinomas in rats with good-to-moderate correlations to an immunohistochemical gold standard. Tumor response biomarkers assessed by dynamic contrast-enhanced computed tomography may be a promising future approach to a more personalized and targeted cancer therapy.
Collapse
Affiliation(s)
- Clemens C. Cyran
- Department of Clinical Radiology, Laboratory for Experimental Radiology, University Hospitals Munich, Grosshadern Campus, Muenchen, Germany
- * E-mail:
| | - Philipp M. Kazmierczak
- Department of Clinical Radiology, Laboratory for Experimental Radiology, University Hospitals Munich, Grosshadern Campus, Muenchen, Germany
| | - Heidrun Hirner
- Department of Clinical Radiology, Laboratory for Experimental Radiology, University Hospitals Munich, Grosshadern Campus, Muenchen, Germany
| | - Matthias Moser
- Department of Clinical Radiology, Laboratory for Experimental Radiology, University Hospitals Munich, Grosshadern Campus, Muenchen, Germany
| | - Michael Ingrisch
- Department of Clinical Radiology, Josef-Lissner-Laboratory for Biomedical Imaging, University Hospitals Munich, Grosshadern Campus, Muenchen, Germany
| | - Lukas Havla
- Department of Clinical Radiology, Josef-Lissner-Laboratory for Biomedical Imaging, University Hospitals Munich, Grosshadern Campus, Muenchen, Germany
| | - Alexandra Michels
- Department of Clinical Radiology, Laboratory for Experimental Radiology, University Hospitals Munich, Grosshadern Campus, Muenchen, Germany
| | - Ralf Eschbach
- Department of Clinical Radiology, Laboratory for Experimental Radiology, University Hospitals Munich, Grosshadern Campus, Muenchen, Germany
| | - Bettina Schwarz
- Department of Surgery, University Hospitals Munich, Grosshadern Campus, Muenchen, Germany
| | - Maximilian F. Reiser
- Department of Clinical Radiology, Laboratory for Experimental Radiology, University Hospitals Munich, Grosshadern Campus, Muenchen, Germany
| | - Christiane J. Bruns
- Department of Surgery, University Hospitals Munich, Grosshadern Campus, Muenchen, Germany
| | - Konstantin Nikolaou
- Department of Clinical Radiology, Laboratory for Experimental Radiology, University Hospitals Munich, Grosshadern Campus, Muenchen, Germany
| |
Collapse
|
30
|
Klarenbeek S, van Miltenburg MH, Jonkers J. Genetically engineered mouse models of PI3K signaling in breast cancer. Mol Oncol 2013; 7:146-64. [PMID: 23478237 DOI: 10.1016/j.molonc.2013.02.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 02/11/2013] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most common type of cancer in women. A substantial fraction of breast cancers have acquired mutations that lead to activation of the phosphoinositide 3-kinase (PI3K) signaling pathway, which plays a central role in cellular processes that are essential in cancer, such as cell survival, growth, division and motility. Oncogenic mutations in the PI3K pathway generally involve either activating mutation of the gene encoding PI3K (PIK3CA) or AKT (AKT1), or loss or reduced expression of PTEN. Several kinases involved in PI3K signaling are being explored as a therapeutic targets for pharmacological inhibition. Despite the availability of a range of inhibitors, acquired resistance may limit the efficacy of single-agent therapy. In this review we discuss the role of PI3K pathway mutations in human breast cancer and relevant genetically engineered mouse models (GEMMs), with special attention to the role of PI3K signaling in oncogenesis, in therapeutic response, and in resistance to therapy. Several sophisticated GEMMs have revealed the cause-and-effect relationships between PI3K pathway mutations and mammary oncogenesis. These GEMMs enable us to study the biology of tumors induced by activated PI3K signaling, as well as preclinical response and resistance to PI3K pathway inhibitors.
Collapse
Affiliation(s)
- Sjoerd Klarenbeek
- Division of Molecular Pathology, Cancer Genomics Centre Netherlands and Cancer Systems Biology Center, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | | | |
Collapse
|
31
|
Dees EC, Carey LA. Improving Endocrine Therapy for Breast Cancer: It's Not That Simple. J Clin Oncol 2013; 31:171-3. [DOI: 10.1200/jco.2012.46.2655] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- E. Claire Dees
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Lisa A. Carey
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|