1
|
Sun Y, Liu L, Zhou L, Yu S, Lan Y, Liang Q, Liu J, Cao A, Liu Y. Tumor Microenvironment-Triggered Charge Reversal Polymetformin-Based Nanosystem Co-Delivered Doxorubicin and IL-12 Cytokine Gene for Chemo-Gene Combination Therapy on Metastatic Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2020; 12:45873-45890. [PMID: 32924511 DOI: 10.1021/acsami.0c14405] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Cancer metastasis is the leading cause of high mortality and disease recurrence in breast cancer. In this study, a novel tumor microenvironment charge reversal polymetformin (PMet)-based nanosystem co-delivering doxorubicin (DOX) and plasmid encoding IL-12 gene (pIL-12) was developed for chemo-gene combination therapy on metastatic breast cancer. Cationic PMet was readily self-assembled into micelles for DOX physical encapsulation and pIL-12 complexation, and a hyaluronidase-sensitive thiolated hyaluronic acid (HA-SH) was then collaboratively assembled to the pIL-12/DOX-PMet micelleplexes, abbreviated as HA/pIL-12/DOX-PMet. DOX/pIL-12 loaded in HA/pIL-12/DOX-PMet micelleplexes presented prolonged circulation in blood, efficient accumulation in tumors, and internalization in tumor cells via CD44 receptor-mediated tumor specific-targeting, and DOX/pIL-12 was co-released in endo/lysosomes tumor microenvironment followed by HAase-triggered HA-SH deshielding from HA/pIL-12/DOX-PMet micelleplexes. Moreover, HA/PMet micelleplexes displayed excellent pIL-12 transfection and IL-12 expression in tumors of 4T1 tumor-bearing mice. Importantly, HA/pIL-12/DOX-PMet micelleplexes synergistically enhanced the NK cells and tumor infiltrated cytotoxic T lymphocytes and modulated the polarization from protumor M2 macrophages to activated antitumor M1 macrophages, with concomitant decreasing of the immunosuppressive regulatory T (Treg) cells, accompanied by an increase in the cytokines expression of IL-12, IFN-γ and TNF-α, consequently showing an improved antitumor and antimetastasis activity in 4T1 breast cancer lung metastasis mice model. In conclusion, the tumor microenvironment charge reversal HA/PMet nanosystem holds great promise for DOX/pIL-12 co-delivery and exploitation in chemo-gene combination therapy on metastatic breast cancer.
Collapse
Affiliation(s)
- Yue Sun
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan 750004, China
| | - Lu Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan 750004, China
| | - Liyue Zhou
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan 750004, China
| | - Shuangyu Yu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan 750004, China
| | - Yang Lan
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan 750004, China
| | - Qiangwei Liang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan 750004, China
| | - Jinxia Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan 750004, China
| | - Aichen Cao
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan 750004, China
| | - Yanhua Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan 750004, China
- Key Laboratory of Hui Ethnic Medicine Modernization, Ningxia Medical University, Yinchuan 750004, China
| |
Collapse
|
2
|
TWIST1 Gene Expression as a Biomarker for Predicting Primary Doxorubicin Resistance in Breast Cancer. Balkan J Med Genet 2019; 22:25-30. [PMID: 31942413 PMCID: PMC6956638 DOI: 10.2478/bjmg-2019-0025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Doxorubicin is one of the most commonly used chemotherapeutic agents for adjuvant chemotherapy of breast cancer. In the studies focused on finding biomarkers to predict the response of the patients and tumors to the drugs used, the Twist transcription factor has been suggested as a candidate biomarker for predicting chemo-resistance of breast tumors. In this study, we aimed to investigate the relationship between TWIST transcription factor expression and the effectiveness of doxorubicin treatment on directly taken primary tumor samples from chemotherapy-naive breast cancer patients. Twenty-six primary breast tumor samples taken from 26 different breast cancer patients were included in this study. Adenosine triphosphate tumor chemo-sensitivity assay (ATP-TCA) has been used to determine tumor response to doxorubicin and real-time reverse-transcription polymerase chain reaction (RT-PCR) was used for analyzing the TWIST1 gene expression of tumors. There was a significant difference in TWIST gene expression between responder and non responder tumors (p <0.05). The TWIST gene expression of the drug-resistant group was higher than the responsive group. This difference was not dependent on the histopathological features of tumors. In conclusion, compatible with earlier studies that have been performed with cell lines, the current study supports the role of higher TWIST gene expression as a biomarker for predicting the response of breast tumors to chemo-therapeutic agent doxorubicin.
Collapse
|
3
|
Flores-Ramírez I, Baranda-Avila N, Langley E. Breast Cancer Stem Cells and Sex Steroid Hormones. Curr Stem Cell Res Ther 2019; 14:398-404. [PMID: 30095060 DOI: 10.2174/1574888x13666180810121415] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 12/14/2022]
Abstract
Breast cancer stem cells (BCSCs) are a small population of tumor-initiating cells that express stem cell-associated markers. In recent years, their properties and mechanisms of regulation have become the focus of intense research due to their intrinsic resistance to conventional cancer therapies. This review describes breast cancer stem cell origin, signaling pathways involved in self-renewal, such as Wnt, Notch and Hedgehog, biomarkers linked to stemness, and the role of sex steroid hormones in BCSC regulation.
Collapse
Affiliation(s)
- Iván Flores-Ramírez
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, CDMX, México.,Departamento de Investigacion Basica, Instituto Nacional de Cancerologia, Av. San Fernando No. 22, Col. Seccion XVI, Tlalpan 14080, CDMX, Mexico
| | - Noemi Baranda-Avila
- Departamento de Investigacion Basica, Instituto Nacional de Cancerologia, Av. San Fernando No. 22, Col. Seccion XVI, Tlalpan 14080, CDMX, Mexico
| | - Elizabeth Langley
- Departamento de Investigacion Basica, Instituto Nacional de Cancerologia, Av. San Fernando No. 22, Col. Seccion XVI, Tlalpan 14080, CDMX, Mexico
| |
Collapse
|
4
|
Batham J, Lim PS, Rao S. SETDB-1: A Potential Epigenetic Regulator in Breast Cancer Metastasis. Cancers (Basel) 2019; 11:cancers11081143. [PMID: 31405032 PMCID: PMC6721492 DOI: 10.3390/cancers11081143] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/05/2019] [Accepted: 08/07/2019] [Indexed: 02/06/2023] Open
Abstract
The full epigenetic repertoire governing breast cancer metastasis is not completely understood. Here, we discuss the histone methyltransferase SET Domain Bifurcated Histone Lysine Methyltransferase 1 (SETDB1) and its role in breast cancer metastasis. SETDB1 serves as an exemplar of the difficulties faced when developing therapies that not only specifically target cancer cells but also the more elusive and aggressive stem cells that contribute to metastasis via epithelial-to-mesenchymal transition and confer resistance to therapies.
Collapse
Affiliation(s)
- Jacob Batham
- Melanie Swan Memorial Translational Centre, Faculty of Sci-Tech, University of Canberra, Bruce ACT 2617, Australia
| | - Pek Siew Lim
- Melanie Swan Memorial Translational Centre, Faculty of Sci-Tech, University of Canberra, Bruce ACT 2617, Australia.
| | - Sudha Rao
- Melanie Swan Memorial Translational Centre, Faculty of Sci-Tech, University of Canberra, Bruce ACT 2617, Australia.
| |
Collapse
|
5
|
Ma F, Liu X, Zhou S, Li W, Liu C, Chadwick M, Qian C. Long non-coding RNA FGF13-AS1 inhibits glycolysis and stemness properties of breast cancer cells through FGF13-AS1/IGF2BPs/Myc feedback loop. Cancer Lett 2019; 450:63-75. [PMID: 30771425 DOI: 10.1016/j.canlet.2019.02.008] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 01/24/2019] [Accepted: 02/10/2019] [Indexed: 12/08/2022]
Abstract
LncRNAs have been proven to play crucial roles in various processes of breast cancer. LncRNA FGF13-AS1 has been identified as one of the 25 downregulated lncRNAs in breast cancer through analyzing data from two cohorts and TCGA by another group of our lab. In this study, we report that FGF13-AS1 expression is decreased in breast cancer tissue compared with corresponding normal tissue, and the downregulation of FGF13-AS1 is associated with poor prognosis. Functional studies show that FGF13-AS1 inhibits breast cancer cells proliferation, migration, and invasion by impairing glycolysis and stemness properties. Mechanistically, FGF13-AS1 reduces the half-life of c-Myc (Myc) mRNA by binding RNA-binding proteins, insulin-like growth factor 2 mRNA binding proteins (IGF2BPs) and disrupting the interaction between IGF2BPs and Myc mRNA. Furthermore, Myc transcriptionally inhibits FGF13-AS1, forming a feedback loop in this signaling pathway. These results reveal for the first time that FGF13-AS1 functions as a tumor suppressor by inhibiting glycolysis and stemness properties of breast cancer cells, and the FGF13-AS1/IGF2BPs/Myc feedback loop could be a novel therapeutic target for breast cancer patients.
Collapse
Affiliation(s)
- Fei Ma
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xu Liu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shibo Zhou
- Department of Pharmacy, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenjie Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Chunxiao Liu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Michelle Chadwick
- Cancer Institute of New Jersey, Rutgers University, New Brunswick, United States
| | - Cheng Qian
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China.
| |
Collapse
|
6
|
Platonov ME, Borovjagin AV, Kaverina N, Xiao T, Kadagidze Z, Lesniak M, Baryshnikova M, Ulasov IV. KISS1 tumor suppressor restricts angiogenesis of breast cancer brain metastases and sensitizes them to oncolytic virotherapy in vitro. Cancer Lett 2017; 417:75-88. [PMID: 29269086 DOI: 10.1016/j.canlet.2017.12.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/13/2017] [Accepted: 12/14/2017] [Indexed: 12/12/2022]
Abstract
KISS1 tumor suppressor protein regulates cancer cell invasion via MMP9 metalloproteinase. Downregulation of KISS1 gene expression promotes progression of breast cancer and melanoma, resulting in the development of distant metastases. In the current study, we investigated whether restoration of KISS1 expression in KISS1-deficient human metastatic breast cancer cells holds potential as an advanced anticancer strategy. To this end we engineered an infectivity-enhanced conditionally-replicative human adenovirus type 5 encoding KISS1 as an "arming" transgene in the Ad5 E3 region for an ectopic KISS1 expression in transduced cancer cells. The oncolytic potential of the vector was examined using brain-invading metastatic clones of CN34 and MDA-MB-231 breast cancer cells, which supported high levels of AdKISS1 replication, correlating with a robust CRAd-mediated cytotoxicity. Secretion of cellular factors responsible for tumor angiogenesis, cell-to-cell communication and anti-tumoral immune responses upon KISS1 expression in breast cancer cells was analyzed by a RayBiotech Kiloplex Quantibody array. Overall, our results indicate that KISS1 transgene expression provides an important benefit for CRAd-mediated cytotoxicity in breast cancer cells and holds potential as an anticancer treatment in conjunction with oncolytic virotherapy of breast and other metastatic cancers.
Collapse
Affiliation(s)
- Mikhail E Platonov
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Anton V Borovjagin
- Institute of Oral Health Research, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Natalya Kaverina
- N.N. Blokhin Cancer Research Center, RAMN, Kashirskoe Shosse 23, Moscow, 115478, Russia
| | - Ting Xiao
- Department of Neurological Surgery, Northwestern University, Chicago, 60611, USA
| | - Zaira Kadagidze
- N.N. Blokhin Cancer Research Center, RAMN, Kashirskoe Shosse 23, Moscow, 115478, Russia
| | - Maciej Lesniak
- Department of Neurological Surgery, Northwestern University, Chicago, 60611, USA
| | - Marya Baryshnikova
- N.N. Blokhin Cancer Research Center, RAMN, Kashirskoe Shosse 23, Moscow, 115478, Russia
| | - Ilya V Ulasov
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, 119991, Russia.
| |
Collapse
|
7
|
Yan Q, Xiaorong Z, Zhang Z, Bing W, Feng Y, Hong B. Prevalence of protein C receptor (PROCR) is associated with inferior clinical outcome in Breast invasive ductal carcinoma. Pathol Res Pract 2017; 213:1173-1179. [PMID: 28756987 DOI: 10.1016/j.prp.2017.06.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 05/23/2017] [Accepted: 06/24/2017] [Indexed: 02/05/2023]
Abstract
Recently, PROCR is reported to play an important role in cell growth, apoptosis, proliferation and tumor relapse. Some researchers thought that PROCR+ cells had cancer stem cell ability, which might contribute to progressive behavior in breast cancer. Our study was to assess the expression of PROCR in invasive ductal carcinoma tissues with their prognostic implications. We enrolled formalin fixed paraffin-embedded tumor tissues of 271 patients diagnosed as invasive ductal breast cancer with clinical stage II or III into our study. Immunohistochemistry staining was performed on all the tissue microarray slides, and result were interpreted by two pathologists with blinded method. We analyzed PROCR expression levels with the clinical characteristics as well as their prognostic values. PROCR expression detected in the cell was interpreted. Chi-square test showed us its positive expression had a close association with distant metastases (p=0.035). Univariate survival analysis indicated that prevalence of PROCR expression in the invasive ductal breast cancer was significantly related with decreased disease-free survival (pDFS=0.010) and overall survival (pOS=0.008). In multivariate survival by Cox proportional hazard model, positive expression group for PROCR was found to have shorter DFS [pDFS=0.028, hazard ratio (95% CI): 1.183(1.069-3.140)]. Our findings suggested that breast cancer patients with expression of PROCR is more prone to suffer from distant metastasis and bad clinical outcomes.
Collapse
Affiliation(s)
- Qiu Yan
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China; Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhong Xiaorong
- Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China; Laboratory of Molecular Diagnosis of Cancer, State Key Laboratory of Biotherapy, National Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhang Zhang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Wei Bing
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ye Feng
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China; Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, PR China
| | - Bu Hong
- Laboratory of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China; Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, PR China; Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
8
|
Yedjou CG, Tchounwou PB, Payton M, Miele L, Fonseca DD, Lowe L, Alo RA. Assessing the Racial and Ethnic Disparities in Breast Cancer Mortality in the United States. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2017; 14:E486. [PMID: 28475137 PMCID: PMC5451937 DOI: 10.3390/ijerph14050486] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 04/22/2017] [Accepted: 04/26/2017] [Indexed: 01/01/2023]
Abstract
Breast cancer is the second leading cause of cancer related deaths among women aged 40-55 in the United States and currently affects more than one in ten women worldwide. It is also one of the most diagnosed cancers in women both in wealthy and poor countries. Fortunately, the mortality rate from breast cancer has decreased in recent years due to increased emphasis on early detection and more effective treatments in White population. Although the mortality rates have declined in some ethnic populations, the overall cancer incidence among African American and Hispanic populations has continued to grow. The goal of the present review article was to highlight similarities and differences in breast cancer morbidity and mortality rates primarily among African American women compared to White women in the United States. To reach our goal, we conducted a search of articles in journals with a primary focus on minority health, and authors who had published articles on racial/ethnic disparity related to breast cancer patients. A systematic search of original research was conducted using MEDLINE, PUBMED and Google Scholar databases. We found that racial/ethnic disparities in breast cancer may be attributed to a large number of clinical and non-clinical risk factors including lack of medical coverage, barriers to early detection and screening, more advanced stage of disease at diagnosis among minorities, and unequal access to improvements in cancer treatment. Many African American women have frequent unknown or unstaged breast cancers than White women. These risk factors may explain the differences in breast cancer treatment and survival rate between African American women and White women. New strategies and approaches are needed to promote breast cancer prevention, improve survival rate, reduce breast cancer mortality, and ultimately improve the health outcomes of racial/ethnic minorities.
Collapse
Affiliation(s)
- Clement G Yedjou
- Natural Chemotherapeutics Research Laboratory, Research Centers in Minority Institutio (RCMI)-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box 18750, Jackson, MS 39217, USA.
| | - Paul B Tchounwou
- Natural Chemotherapeutics Research Laboratory, Research Centers in Minority Institutio (RCMI)-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box 18750, Jackson, MS 39217, USA.
| | - Marinelle Payton
- Center of Excellence in Minority Health and Health Disparities, School of Public Health, Jackson State University, Jackson Medical Mall-Thad Cochran Center, 350 West Woodrow Wilson Avenue, Jackson, MS 39213, USA.
| | - Lucio Miele
- Department of Genetics, LSU Health Sciences Center, School of Medicine, 533 Bolivar Street, Room 657, New Orleans, LA 70112, USA.
| | - Duber D Fonseca
- Natural Chemotherapeutics Research Laboratory, Research Centers in Minority Institutio (RCMI)-Center for Environmental Health, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box 18750, Jackson, MS 39217, USA.
| | - Leroy Lowe
- Lancaster Environment Centre, Lancaster University, Bailrigg, Lancaster LA1 4YW, UK.
| | - Richard A Alo
- Department of Civil and Environmental Engineering, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch Street, Box 18750, Jackson, MS 39217, USA.
| |
Collapse
|
9
|
Zhang Z, Liu L, Cao S, Zhu Y, Mei Q. Gene delivery of TIPE2 inhibits breast cancer development and metastasis via CD8 + T and NK cell-mediated antitumor responses. Mol Immunol 2017; 85:230-237. [PMID: 28314212 DOI: 10.1016/j.molimm.2017.03.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 03/06/2017] [Accepted: 03/06/2017] [Indexed: 10/20/2022]
Abstract
Breast cancer is the second leading cause of cancer-related deaths in the female patients which was mainly caused by metastasis. Development of target gene therapy for breast cancer to suppress tumor progress and metastasis will improve the therapeutic options and be of great benefit to the patients. Tumor necrosis factor-alpha-induced protein 8-like 2 is a novel molecule for maintaining immune homeostasis and is involved in cancer development. In the present study, we overexpressed TIPE2 in breast cancer cells to investigate the role of TIPE2 in the development of breast cancer. Our results showed that overexpression of TIPE2 significantly inhibited the proliferation of 4T1 cells in vitro and in vivo. We constructed a non-viral targeted gene therapeutic system by using the minicircle plasmids expressing TIPE2. We found that the growth and metastasis of breast cancer was significantly inhibited by hydrodynamic gene delivery of TIPE2 plasmids in vivo. Mechanistically, TIPE2 increased T and NK cells, and decreased MDSCs. Gene delivery of TIPE2 up-regulated the production of IFN-γ and TNF-α by CD8+ T and NK cells in spleens and tumor microenvironment, and enhanced the cytotoxic activity of CD8+ T and NK cells. Taken together, TIPE2 inhibited breast cancer development and metastasis possibly via promoting CD8+ T and NK cell-mediated antitumor immune responses. Thus, the results indicate that TIPE2 may be a potential therapeutic target for breast cancer therapy.
Collapse
Affiliation(s)
- Zhenhua Zhang
- School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China; State Institute of Pharmaceutical Industry, Shanghai 201203, People's Republic of China
| | - Li Liu
- State Institute of Pharmaceutical Industry, Shanghai 201203, People's Republic of China
| | - Shousong Cao
- State Institute of Pharmaceutical Industry, Shanghai 201203, People's Republic of China
| | - Yizhun Zhu
- School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China.
| | - Qibing Mei
- State Institute of Pharmaceutical Industry, Shanghai 201203, People's Republic of China.
| |
Collapse
|
10
|
Hu H, Sun J, Wang C, Bu X, Liu X, Mao Y, Wang H. IL-33 facilitates endocrine resistance of breast cancer by inducing cancer stem cell properties. Biochem Biophys Res Commun 2017; 485:643-650. [PMID: 28216163 DOI: 10.1016/j.bbrc.2017.02.080] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 02/14/2017] [Indexed: 02/08/2023]
Abstract
Breast cancers with estrogen receptor (ER) expressions account for the majority of all clinical cases. Due to hormone therapy with tamoxifen, prognoses of patients with ER-positive breast cancer are significantly improved. However, endocrine resistance to tamoxifen is common and inevitable, leading to compromised efficacy of hormone therapy. Herein, we identify a crucial role of IL-33 in inducing endocrine resistance of breast cancer. IL-33 overexpression in breast cancer cells results in resistance to tamoxifen-induced tumor growth inhibition, while IL-33 knockdown corrects this problem. Mechanistically, IL-33 induces breast cancer stem cell properties evidenced by mammosphere formation and xenograft tumorigenesis, as well as expression of cancer stem cell genes including ALDH1A3, OCT4, NANOG and SOX2. In breast cancer patients, higher serum IL-33 levels portend advanced clinical stages, poorly differentiated cancer cells and tumor recurrence. IL-33 expression levels in patients' freshly isolated breast cancer cells predicts tamoxifen resistance and cancer stem cell features in individual patient. Collectively, IL-33 induces endocrine resistance of breast cancer by promoting cancer stem cell properties. These findings provide novel mechanisms connecting IL-33 with cancer pathogenesis and pinpoint IL-33 as a promising target for optimizing hormone therapy in clinical practice.
Collapse
Affiliation(s)
- Haiyan Hu
- Center of Diagnosis and Treatment of Breast Disease, The Affiliated Hospital of Qingdao University, Qingdao 266071, China
| | - Jiaxing Sun
- Department of Respiratory Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266071, China
| | - Chunhong Wang
- Department of Respiratory Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xiangmao Bu
- Department of Clinical Laboratory, Qingdao Women & Children Hospital, Qingdao 266034, China
| | - Xiangping Liu
- Central Laboratory of Molecular Biology, The Affiliated Hospital of Qingdao University, Qingdao 266071, China
| | - Yan Mao
- Center of Diagnosis and Treatment of Breast Disease, The Affiliated Hospital of Qingdao University, Qingdao 266071, China
| | - Haibo Wang
- Center of Diagnosis and Treatment of Breast Disease, The Affiliated Hospital of Qingdao University, Qingdao 266071, China.
| |
Collapse
|
11
|
Peng F, Li TT, Wang KL, Xiao GQ, Wang JH, Zhao HD, Kang ZJ, Fan WJ, Zhu LL, Li M, Cui B, Zheng FM, Wang HJ, Lam EWF, Wang B, Xu J, Liu Q. H19/let-7/LIN28 reciprocal negative regulatory circuit promotes breast cancer stem cell maintenance. Cell Death Dis 2017; 8:e2569. [PMID: 28102845 PMCID: PMC5386357 DOI: 10.1038/cddis.2016.438] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 11/14/2016] [Accepted: 11/25/2016] [Indexed: 12/12/2022]
Abstract
Long noncoding RNA-H19 (H19), an imprinted oncofetal gene, has a central role in carcinogenesis. Hitherto, the mechanism by which H19 regulates cancer stem cells, remains elusive. Here we show that breast cancer stem cells (BCSCs) express high levels of H19, and ectopic overexpression of H19 significantly promotes breast cancer cell clonogenicity, migration and mammosphere-forming ability. Conversely, silencing of H19 represses these BCSC properties. In concordance, knockdown of H19 markedly inhibits tumor growth and suppresses tumorigenesis in nude mice. Mechanistically, we found that H19 functions as a competing endogenous RNA to sponge miRNA let-7, leading to an increase in expression of a let-7 target, the core pluripotency factor LIN28, which is enriched in BCSC populations and breast patient samples. Intriguingly, this gain of LIN28 expression can also feedback to reverse the H19 loss-mediated suppression of BCSC properties. Our data also reveal that LIN28 blocks mature let-7 production and, thereby, de-represses H19 expression in breast cancer cells. Appropriately, H19 and LIN28 expression exhibits strong correlations in primary breast carcinomas. Collectively, these findings reveal that lncRNA H19, miRNA let-7 and transcriptional factor LIN28 form a double-negative feedback loop, which has a critical role in the maintenance of BCSCs. Consequently, disrupting this pathway provides a novel therapeutic strategy for breast cancer.
Collapse
Affiliation(s)
- Fei Peng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian; State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Ting-Ting Li
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian; State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Kai-Li Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian; State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Guo-Qing Xiao
- Department of Breast Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Ju-Hong Wang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Hai-Dong Zhao
- Department of Breast Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Zhi-Jie Kang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian; State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou 510060, China
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Wen-Jun Fan
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian; State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Li-Li Zhu
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Mei Li
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Bai Cui
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian; State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Fei-Meng Zheng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian; State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou 510060, China
- Department of Medical Oncology, The Eastern Hospital of The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510700, China
| | - Hong-Jiang Wang
- Department of Breast Surgery, The First Affiliated Hospital, Dalian Medical University, Dalian 116011, China
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK
| | - Bo Wang
- Department of Medical Oncology, The Eastern Hospital of The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510700, China
| | - Jie Xu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian; State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Quentin Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian; State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou 510060, China
| |
Collapse
|
12
|
Xu LZ, Li SS, Zhou W, Kang ZJ, Zhang QX, Kamran M, Xu J, Liang DP, Wang CL, Hou ZJ, Wan XB, Wang HJ, Lam EWF, Zhao ZW, Liu Q. p62/SQSTM1 enhances breast cancer stem-like properties by stabilizing MYC mRNA. Oncogene 2017; 36:304-317. [PMID: 27345399 PMCID: PMC5269535 DOI: 10.1038/onc.2016.202] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 02/04/2016] [Accepted: 04/08/2016] [Indexed: 12/12/2022]
Abstract
Aberrant p62 overexpression has been implicated in breast cancer development. Here, we found that p62 expression was elevated in breast cancer stem cells (BCSCs), including CD44+CD24- fractions, mammospheres, ALDH1+ populations and side population cells. Indeed, short-hairpin RNA (shRNA)-mediated knockdown of p62 impaired breast cancer cells from self-renewing under anchorage-independent conditions, whereas ectopic overexpression of p62 enhanced the self-renewal ability of breast cancer cells in vitro. Genetic depletion of p62 robustly inhibited tumor-initiating frequencies, as well as growth rates of BCSC-derived tumor xenografts in immunodeficient mice. Consistently, immunohistochemical analysis of clinical breast tumor tissues showed that high p62 expression levels were linked to poorer clinical outcome. Further gene expression profiling analysis revealed that p62 was positively correlated with MYC expression level, which mediated the function of p62 in promoting breast cancer stem-like properties. MYC mRNA level was reduced upon p62 deletion by siRNA and increased with p62 overexpression in breast cancer cells, suggesting that p62 positively regulated MYC mRNA. Interestingly, p62 did not transactivate MYC promoter. Instead, p62 delayed the degradation of MYC mRNA by repressing the expression of let-7a and let-7b, thus promoting MYC mRNA stabilization at the post-transcriptional level. Consistently, let-7a and let-7b mimics attenuated p62-mediated MYC mRNA stabilization. Together, these findings unveiled a previously unappreciated role of p62 in the regulation of BCSCs, assigning p62 as a promising therapeutic target for breast cancer treatments.
Collapse
Affiliation(s)
- L-Z Xu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - S-S Li
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - W Zhou
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Z-J Kang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Q-X Zhang
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - M Kamran
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - J Xu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - D-P Liang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - C-L Wang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Z-J Hou
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - X-B Wan
- Department of Radiation Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - H-J Wang
- Department of Breast Surgery, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - E W-F Lam
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Z-W Zhao
- Department of Breast Surgery, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
- Department of Breast Surgery, The Second Affiliated Hospital, Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian, Liaoning 116000, China. E-mail:
| | - Q Liu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian, Liaoning 116044, China E-mail:
| |
Collapse
|
13
|
Zhang Z, Liu L, Liu C, Cao S, Zhu Y, Mei Q. TIPE2 suppresses the tumorigenesis, growth and metastasis of breast cancer via inhibition of the AKT and p38 signaling pathways. Oncol Rep 2016; 36:3311-3316. [PMID: 27779698 DOI: 10.3892/or.2016.5192] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 06/21/2016] [Indexed: 11/06/2022] Open
|
14
|
Fiorillo M, Verre AF, Iliut M, Peiris-Pagés M, Ozsvari B, Gandara R, Cappello AR, Sotgia F, Vijayaraghavan A, Lisanti MP. Graphene oxide selectively targets cancer stem cells, across multiple tumor types: implications for non-toxic cancer treatment, via "differentiation-based nano-therapy". Oncotarget 2016; 6:3553-62. [PMID: 25708684 PMCID: PMC4414136 DOI: 10.18632/oncotarget.3348] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Accepted: 02/12/2015] [Indexed: 11/25/2022] Open
Abstract
Tumor-initiating cells (TICs), a.k.a. cancer stem cells (CSCs), are difficult to eradicate with conventional approaches to cancer treatment, such as chemo-therapy and radiation. As a consequence, the survival of residual CSCs is thought to drive the onset of tumor recurrence, distant metastasis, and drug-resistance, which is a significant clinical problem for the effective treatment of cancer. Thus, novel approaches to cancer therapy are needed urgently, to address this clinical need. Towards this end, here we have investigated the therapeutic potential of graphene oxide to target cancer stem cells. Graphene and its derivatives are well-known, relatively inert and potentially non-toxic nano-materials that form stable dispersions in a variety of solvents. Here, we show that graphene oxide (of both big and small flake sizes) can be used to selectively inhibit the proliferative expansion of cancer stem cells, across multiple tumor types. For this purpose, we employed the tumor-sphere assay, which functionally measures the clonal expansion of single cancer stem cells under anchorage-independent conditions. More specifically, we show that graphene oxide effectively inhibits tumor-sphere formation in multiple cell lines, across 6 different cancer types, including breast, ovarian, prostate, lung and pancreatic cancers, as well as glioblastoma (brain). In striking contrast, graphene oxide is non-toxic for "bulk" cancer cells (non-stem) and normal fibroblasts. Mechanistically, we present evidence that GO exerts its striking effects on CSCs by inhibiting several key signal transduction pathways (WNT, Notch and STAT-signaling) and thereby inducing CSC differentiation. Thus, graphene oxide may be an effective non-toxic therapeutic strategy for the eradication of cancer stem cells, via differentiation-based nano-therapy.
Collapse
Affiliation(s)
- Marco Fiorillo
- The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, University of Manchester, UK.,The Breakthrough Breast Cancer Research Unit, Institute of Cancer Sciences, University of Manchester, UK.,The Department of Pharmacy, Health and Nutritional Sciences, The University of Calabria, Italy
| | - Andrea F Verre
- School of Materials and National Graphene Institute, University of Manchester, UK
| | - Maria Iliut
- School of Materials and National Graphene Institute, University of Manchester, UK
| | - Maria Peiris-Pagés
- The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, University of Manchester, UK.,The Breakthrough Breast Cancer Research Unit, Institute of Cancer Sciences, University of Manchester, UK
| | - Bela Ozsvari
- The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, University of Manchester, UK.,The Breakthrough Breast Cancer Research Unit, Institute of Cancer Sciences, University of Manchester, UK
| | - Ricardo Gandara
- The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, University of Manchester, UK.,The Breakthrough Breast Cancer Research Unit, Institute of Cancer Sciences, University of Manchester, UK
| | - Anna Rita Cappello
- The Department of Pharmacy, Health and Nutritional Sciences, The University of Calabria, Italy
| | - Federica Sotgia
- The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, University of Manchester, UK.,The Breakthrough Breast Cancer Research Unit, Institute of Cancer Sciences, University of Manchester, UK
| | | | - Michael P Lisanti
- The Manchester Centre for Cellular Metabolism (MCCM), Institute of Cancer Sciences, University of Manchester, UK.,The Breakthrough Breast Cancer Research Unit, Institute of Cancer Sciences, University of Manchester, UK
| |
Collapse
|
15
|
Pei S, Yang X, Wang H, Zhang H, Zhou B, Zhang D, Lin D. Plantamajoside, a potential anti-tumor herbal medicine inhibits breast cancer growth and pulmonary metastasis by decreasing the activity of matrix metalloproteinase-9 and -2. BMC Cancer 2015; 15:965. [PMID: 26674531 PMCID: PMC4682252 DOI: 10.1186/s12885-015-1960-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 11/30/2015] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Metastasis is the major cause of death in breast cancers. MMPs play a key role in tumor microenvironment that facilitates metastasis. The existing researches suggest that the high expression of gelatinase A and B (MMP2 and MMP9) promote the metastasis of breast cancer. Therefore, gelatinase inhibitor can effectively suppress tumor metastasis. However, at present, there is no dramatically effective gelatinase inhibitor against breast cancer. METHODS We screened gelatinase inhibitor among Chinese herbal medicine by molecular docking technology; investigated the proliferation, migration and invasion of MDA-MB-231 human breast cancer cell line and 4T1 mouse breast cancer cell line in response to the treatment with the screened inhibitor by wound assay, invasion assay and gelatin zymography; then further examined the effects of inhibitor on allograft mammary tumors of mice by immunohistochemistry. RESULTS We successfully screened an Chinese herbal medicine-Plantamajoside(PMS)-which can reduce the gelatinase activity of MMP9 and MMP2. In vitro, PMS can inhibit the proliferation, migration and invasion of MDA-MB-231 human breast cancer cell line and 4T1 mouse breast cancer cell line by decreasing MMP9 and MMP2 activity. In vivo, oral administration of PMS to the mice bearing 4T1 cells induced tumors resulted in significant reduction in allograft tumor volume and weights, significant decrease in microvascular density and significant lower lung metastasis rate. CONCLUSIONS Our results indicate that as a promising anti-cancer agent, PMS may inhibit growth and metastasis of breast cancer by inhibiting the activity of MMP9 and MMP2.
Collapse
Affiliation(s)
- Shimin Pei
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| | - Xu Yang
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| | - Huanan Wang
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Hong Zhang
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| | - Bin Zhou
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| | - Di Zhang
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| | - Degui Lin
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
16
|
Mohammad RM, Muqbil I, Lowe L, Yedjou C, Hsu HY, Lin LT, Siegelin MD, Fimognari C, Kumar NB, Dou QP, Yang H, Samadi AK, Russo GL, Spagnuolo C, Ray SK, Chakrabarti M, Morre JD, Coley HM, Honoki K, Fujii H, Georgakilas AG, Amedei A, Niccolai E, Amin A, Ashraf SS, Helferich WG, Yang X, Boosani CS, Guha G, Bhakta D, Ciriolo MR, Aquilano K, Chen S, Mohammed SI, Keith WN, Bilsland A, Halicka D, Nowsheen S, Azmi AS. Broad targeting of resistance to apoptosis in cancer. Semin Cancer Biol 2015; 35 Suppl:S78-S103. [PMID: 25936818 PMCID: PMC4720504 DOI: 10.1016/j.semcancer.2015.03.001] [Citation(s) in RCA: 596] [Impact Index Per Article: 59.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 03/04/2015] [Accepted: 03/04/2015] [Indexed: 12/15/2022]
Abstract
Apoptosis or programmed cell death is natural way of removing aged cells from the body. Most of the anti-cancer therapies trigger apoptosis induction and related cell death networks to eliminate malignant cells. However, in cancer, de-regulated apoptotic signaling, particularly the activation of an anti-apoptotic systems, allows cancer cells to escape this program leading to uncontrolled proliferation resulting in tumor survival, therapeutic resistance and recurrence of cancer. This resistance is a complicated phenomenon that emanates from the interactions of various molecules and signaling pathways. In this comprehensive review we discuss the various factors contributing to apoptosis resistance in cancers. The key resistance targets that are discussed include (1) Bcl-2 and Mcl-1 proteins; (2) autophagy processes; (3) necrosis and necroptosis; (4) heat shock protein signaling; (5) the proteasome pathway; (6) epigenetic mechanisms; and (7) aberrant nuclear export signaling. The shortcomings of current therapeutic modalities are highlighted and a broad spectrum strategy using approaches including (a) gossypol; (b) epigallocatechin-3-gallate; (c) UMI-77 (d) triptolide and (e) selinexor that can be used to overcome cell death resistance is presented. This review provides a roadmap for the design of successful anti-cancer strategies that overcome resistance to apoptosis for better therapeutic outcome in patients with cancer.
Collapse
Affiliation(s)
- Ramzi M Mohammad
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States; Interim translational Research Institute, Hamad Medical Corporation, Doha, Qatar.
| | - Irfana Muqbil
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Leroy Lowe
- Getting to Know Cancer, Truro, Nova Scotia, Canada
| | - Clement Yedjou
- C-SET, [Jackson, #229] State University, Jackson, MS, United States
| | - Hsue-Yin Hsu
- Department of Life Sciences, Tzu-Chi University, Hualien, Taiwan
| | - Liang-Tzung Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Markus David Siegelin
- Department of Pathology and Cell Biology, Columbia University, New York City, NY, United States
| | - Carmela Fimognari
- Dipartimento di Scienze per la Qualità della Vita Alma Mater Studiorum-Università di Bologna, Italy
| | - Nagi B Kumar
- Moffit Cancer Center, University of South Florida College of Medicine, Tampa, FL, United States
| | - Q Ping Dou
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States; Departments of Pharmacology and Pathology, Karmanos Cancer Institute, Detroit MI, United States
| | - Huanjie Yang
- The School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | | | - Gian Luigi Russo
- Institute of Food Sciences National Research Council, Avellino, Italy
| | - Carmela Spagnuolo
- Institute of Food Sciences National Research Council, Avellino, Italy
| | - Swapan K Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Mrinmay Chakrabarti
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| | - James D Morre
- Mor-NuCo, Inc, Purdue Research Park, West Lafayette, IN, United States
| | - Helen M Coley
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, United Kingdom
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Japan
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Japan
| | - Alexandros G Georgakilas
- Department of Physics, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Zografou 15780, Athens, Greece
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, university of florence, Italy
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, university of florence, Italy
| | - Amr Amin
- Department of Biology, College of Science, UAE University, United Arab Emirates; Faculty of Science, Cairo University, Egypt
| | - S Salman Ashraf
- Department of Chemistry, College of Science, UAE University, United Arab Emirates
| | - William G Helferich
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Xujuan Yang
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Chandra S Boosani
- Department of BioMedical Sciences, School of Medicine Creighton University, Omaha NE, United States
| | - Gunjan Guha
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | - Dipita Bhakta
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | | | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", Italy
| | - Sophie Chen
- Ovarian and Prostate Cancer Research Trust Laboratory, Guildford, Surrey, United Kingdom
| | - Sulma I Mohammed
- Department of Comparative Pathobiology and Purdue University Center for Cancer Research, Purdue, West Lafayette, IN, United States
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Ireland
| | - Alan Bilsland
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Ireland
| | - Dorota Halicka
- Department of Pathology, New York Medical College, Valhalla, NY, United States
| | - Somaira Nowsheen
- Mayo Graduate School, Mayo Medical School, Mayo Clinic Medical Scientist Training Program, Rochester, MN, United States
| | - Asfar S Azmi
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| |
Collapse
|
17
|
Pu H, Zhang Q, Zhao C, Shi L, Wang Y, Wang J, Zhang M. Overexpression of G6PD is associated with high risks of recurrent metastasis and poor progression-free survival in primary breast carcinoma. World J Surg Oncol 2015; 13:323. [PMID: 26607846 PMCID: PMC4660828 DOI: 10.1186/s12957-015-0733-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 11/10/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The present study aimed to investigate the expression of CYP27A1, CYP7B1, insulin-like growth factor-1 (IGF-1), glucose-6-phosphate-dehydrogenase (G6PD), glutathione S-transferase P1 (GSTP1), and pyruvate kinase isoform M2 (PKM2) in breast carcinoma tissue and evaluate their prognostic value for progression-free survival (PFS) and overall survival (OS). METHODS A total of 20 patients treated with surgery for primary breast carcinoma were enrolled: 10 cases diagnosed with recurrent metastasis (A), along with their corresponding metastases specimen (AM) and 10 cases with no evidence of recurrence or metastasis (B). Baseline characteristics of patients including age, lymph node metastasis, molecular subtypes, tumor staging and size, and pathological classification were all collected. Immunohistochemistry was performed to detect the protein expression in tumor specimens. RESULTS Elevated G6PD protein levels were noted in group A compared with group AM and B (both P < 0.05), and PKM2 expression was also higher in group A when compared to group AM (P = 0.019), but similar with group B (P > 0.05). No association between clinicopathological parameters and the two proteins expression was observed. The G6PD protein expression was strongly associated with PFS of breast carcinoma patients (P = 0.021) but not for OS. According to the Kaplan-Meier analysis, mean PFS time of patients with G6PD-negative and G6PD-positive expression tumor were 71.36 ± 6.53 and 32.25 ± 5.67 months, respectively (P = 0.002). CONCLUSIONS The G6PD protein could be served as a potential prognostic biomarker for primary breast carcinoma, and overexpression of G6PD protein predicted a high risk of recurrent metastasis and poor PFS during follow-up.
Collapse
MESH Headings
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/enzymology
- Breast Neoplasms/pathology
- Breast Neoplasms/surgery
- Carcinoma, Ductal, Breast/enzymology
- Carcinoma, Ductal, Breast/secondary
- Carcinoma, Ductal, Breast/surgery
- Carrier Proteins/metabolism
- Female
- Follow-Up Studies
- Glucosephosphate Dehydrogenase/metabolism
- Glutathione S-Transferase pi/metabolism
- Humans
- Immunoenzyme Techniques
- Lymphatic Metastasis
- Membrane Proteins/metabolism
- Middle Aged
- Neoplasm Grading
- Neoplasm Invasiveness
- Neoplasm Recurrence, Local/diagnosis
- Neoplasm Recurrence, Local/enzymology
- Neoplasm Recurrence, Local/surgery
- Neoplasm Staging
- Prognosis
- Survival Rate
- Thyroid Hormones/metabolism
- Thyroid Hormone-Binding Proteins
Collapse
Affiliation(s)
- Haihong Pu
- Department of Medical Oncology, The Third Affiliated Hospital of Harbin Medical University, Haping Road 150 of Nangang District, Harbin, Heilongjiang Province, 150086, China.
| | - Qingyuan Zhang
- Department of Medical Oncology, The Third Affiliated Hospital of Harbin Medical University, Haping Road 150 of Nangang District, Harbin, Heilongjiang Province, 150086, China.
| | - Chunbo Zhao
- Department of Medical Oncology, The Third Affiliated Hospital of Harbin Medical University, Haping Road 150 of Nangang District, Harbin, Heilongjiang Province, 150086, China.
| | - Lei Shi
- Department of radiation oncology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, China.
| | - Yan Wang
- Department of Medical Oncology, The Third Affiliated Hospital of Harbin Medical University, Haping Road 150 of Nangang District, Harbin, Heilongjiang Province, 150086, China.
| | - Jingxuan Wang
- Department of Medical Oncology, The Third Affiliated Hospital of Harbin Medical University, Haping Road 150 of Nangang District, Harbin, Heilongjiang Province, 150086, China.
| | - Minghui Zhang
- Department of Medical Oncology, The Third Affiliated Hospital of Harbin Medical University, Haping Road 150 of Nangang District, Harbin, Heilongjiang Province, 150086, China.
| |
Collapse
|
18
|
Kiyomi A, Makita M, Ozeki T, Li N, Satomura A, Tanaka S, Onda K, Sugiyama K, Iwase T, Hirano T. Characterization and Clinical Implication of Th1/Th2/Th17 Cytokines Produced from Three-Dimensionally Cultured Tumor Tissues Resected from Breast Cancer Patients. Transl Oncol 2015; 8:318-26. [PMID: 26310378 PMCID: PMC4562984 DOI: 10.1016/j.tranon.2015.06.004] [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: 04/09/2015] [Revised: 06/17/2015] [Accepted: 06/23/2015] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES: Several cytokines secreted from breast cancer tissues are suggested to be related to disease prognosis. We examined Th1/Th2/Th17 cytokines produced from three-dimensionally cultured breast cancer tissues and related them with patient clinical profiles. METHODS: 21 tumor tissues and 9 normal tissues surgically resected from breast cancer patients were cultured in thermoreversible gelatin polymer–containing medium. Tissue growth and Th1/Th2/Th17 cytokine concentrations in the culture medium were analyzed and were related with hormone receptor expressions and patient clinical profiles. RESULTS: IL-6 and IL-10 were expressed highly in culture medium of both cancer and normal tissues. However, IFN-γ, TNF-α, IL-2, and IL-17A were not detected in the supernatant of the three-dimensionally cultured normal mammary gland and are seemed to be specific to breast cancer tissues. The growth abilities of hormone receptor–negative cancer tissues were significantly higher than those of receptor-positive tissues (P = 0.0383). Cancer tissues of stage ≥ IIB patients expressed significantly higher TNF-α levels as compared with those of patients with stage < IIB (P = 0.0096). CONCLUSIONS: The tumor tissues resected from breast cancer patients can grow in the three-dimensional thermoreversible gelatin polymer culture system and produce Th1/Th2/Th17 cytokines. Hormone receptor–positive cancer tissues showed less growth ability. TNF-α is suggested to be a biomarker for the cancer stage.
Collapse
Affiliation(s)
- Anna Kiyomi
- Tokyo University of Pharmacy and Life Sciences, Department of Clinical Pharmacology, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan.
| | - Masujiro Makita
- The Cancer Institute Hospital of JFCR, Department of Breast Oncology, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan; Nippon Medical School Musashi Kosugi Hospital, Department of Breast Surgery, 1-396 Kosugimachi, Nakahara-ku, Kawasaki, Kanagawa 211-8533, Japan.
| | - Tomoko Ozeki
- Tokyo University of Pharmacy and Life Sciences, Department of Clinical Pharmacology, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan.
| | - Na Li
- Tokyo University of Pharmacy and Life Sciences, Department of Clinical Pharmacology, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan.
| | - Aiko Satomura
- Tokyo University of Pharmacy and Life Sciences, Department of Clinical Pharmacology, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan.
| | - Sachiko Tanaka
- Tokyo University of Pharmacy and Life Sciences, Department of Clinical Pharmacology, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan.
| | - Kenji Onda
- Tokyo University of Pharmacy and Life Sciences, Department of Clinical Pharmacology, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan.
| | - Kentaro Sugiyama
- Tokyo University of Pharmacy and Life Sciences, Department of Clinical Pharmacology, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan.
| | - Takuji Iwase
- The Cancer Institute Hospital of JFCR, Department of Breast Oncology, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan.
| | - Toshihiko Hirano
- Tokyo University of Pharmacy and Life Sciences, Department of Clinical Pharmacology, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan.
| |
Collapse
|
19
|
YU SHIMIN, HU DONGHUI, ZHANG JIANJUN. Umbelliferone exhibits anticancer activity via the induction of apoptosis and cell cycle arrest in HepG2 hepatocellular carcinoma cells. Mol Med Rep 2015; 12:3869-3873. [DOI: 10.3892/mmr.2015.3797] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 02/11/2015] [Indexed: 11/05/2022] Open
|