1
|
Flont M, Jastrzębska E. A Multi-Layer Breast Cancer Model to Study the Synergistic Effect of Photochemotherapy. MICROMACHINES 2023; 14:1806. [PMID: 37763969 PMCID: PMC10535669 DOI: 10.3390/mi14091806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023]
Abstract
Breast cancer is one of the most common cancers among women. The development of new and effective therapeutic approaches in the treatment of breast cancer is an important challenge in modern oncology. Two-dimensional (2D) cell cultures are most often used in the study of compounds with potential anti-tumor nature. However, it is necessary to develop advanced three-dimensional (3D) cell models that can, to some extent, reflect the physiological conditions. The use of miniature cancer-on-a-chip microfluidic systems can help to mimic the complex cancer microenvironment. In this report, we developed a 3D breast cancer model in the form of a cell multilayer, composed of stromal cells (HMF) and breast cancer parenchyma (MCF-7). The developed cell model was successfully used to analyze the effectiveness of combined sequential photochemotherapy, based on doxorubicin and meso-tetraphenylporphyrin. We proved that the key factor that allows achieving the synergistic effect of combination therapy are the order of drug administration to the cells and the sequence of therapeutic procedures. To the best of our knowledge, studies on the effectiveness of combination photochemotherapy depending on the sequence of the component drugs were performed for the first time under microfluidic conditions on a 3D multilayered model of breast cancer tissue.
Collapse
Affiliation(s)
- Magdalena Flont
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland;
- Center for Advanced Materials and Technologies CEZAMAT, Warsaw University of Technology, Poleczki 19, 02-822 Warsaw, Poland
| | - Elżbieta Jastrzębska
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland;
- Center for Advanced Materials and Technologies CEZAMAT, Warsaw University of Technology, Poleczki 19, 02-822 Warsaw, Poland
| |
Collapse
|
2
|
Iqubal MK, Kaur H, Md S, Alhakamy NA, Iqubal A, Ali J, Baboota S. A technical note on emerging combination approach involved in the onconanotherapeutics. Drug Deliv 2022; 29:3197-3212. [PMID: 36226570 PMCID: PMC9578464 DOI: 10.1080/10717544.2022.2132018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cancer is the second cause of mortality worldwide, and the currently available conventional treatment approach is associated with serious side effects and poor clinical outcomes. Based on the outcome of the exploratory preclinical and clinical studies, it was found that therapeutic response increases multiple folds when anticancer drugs are used in combination. However, the conventional combination of anticancer drugs was associated with various limitations such as increased cost of treatment, systemic toxicity, drug resistance, and reduced pharmacokinetic attributes. Hence, attempts were made to formulate nanocarrier fabricated combinatorial drugs (NFCDs) to effectively manage and treat cancer. This approach offers several advantages, such as improved stability, lower drug exposure, targeted drug delivery, low side effects, and improved clinical outcome. Hence, in this review, first time, we have discussed the recent advancement and various types of nano carrier-based combinatorial drug delivery systems in a different type of cancer and highlighted the personalized combinatorial theranostic medicine as a futuristic anticancer treatment approach.
Collapse
Affiliation(s)
- Mohammad Kashif Iqubal
- Product Development Department, Sentiss Research Centre, Sentiss Pharma Pvt Ltd, Gurugram, India.,Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Harsimran Kaur
- Department of Pharmaceutics, Delhi Pharmaceutical Science and Research University, New Delhi, India
| | - Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nabil A Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Sanjula Baboota
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| |
Collapse
|
3
|
Passos Gibson V, Derbali RM, Phan HT, Tahiri H, Allen C, Hardy P, Chain JL. Survivin silencing improved the cytotoxicity of carboplatin and melphalan in Y79 and primary retinoblastoma cells. Int J Pharm 2020; 589:119824. [PMID: 32861768 DOI: 10.1016/j.ijpharm.2020.119824] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/11/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022]
Abstract
Survivin stands out as one of the most specific cancer targets discovered to date. Although single inhibition, e.g. through small interfering RNA (siRNA), has shown modest results in clinical trials, its combination with drugs holds promise to sensitize cancer cells to chemotherapeutics. In this study, we propose a sequential treatment of siRNA survivin followed by chemotherapy. Firstly, we demonstrated that siRNA-loaded switchable lipid nanoparticles (siLNP) silence survivin in a panel of cancer cell lines. Subsequently, we selected retinoblastoma (RB) as our model to screen four chemotherapeutic agents: carboplatin, topotecan, melphalan or teniposide. The effect of drugs on survivin expression and caspase-3 was investigated by RT-qPCR. The best drug combination was selected measuring the viability, survivin expression and the selectivity of the treatment. Our stepwise method revealed that siRNA delivery by switchable LNP sensitized Y79, but not the healthy APRE-19 cell line, to carboplatin and melphalan cytotoxicity. This ability was validated on primary human RB cells. Finally, the distinct behavior of the drugs demonstrated that a diligent screening of drugs should be envisioned when looking for synergy with survivin. Our sequential approach highlighted carboplatin and melphalan as agents to be investigated in future survivin-associated in vivo testing to tackle RB.
Collapse
Affiliation(s)
- Victor Passos Gibson
- Gene Delivery Laboratory, Faculty of Pharmacy, Université de Montréal, H3C 3J7 Montréal, Québec, Canada
| | - Rabeb Mouna Derbali
- Gene Delivery Laboratory, Faculty of Pharmacy, Université de Montréal, H3C 3J7 Montréal, Québec, Canada
| | - Huu Trong Phan
- Gene Delivery Laboratory, Faculty of Pharmacy, Université de Montréal, H3C 3J7 Montréal, Québec, Canada
| | - Houda Tahiri
- Department of Pediatrics, Physiology and Pharmacology, Université de Montréal, H3C 3J7 Montréal, Québec, Canada
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Pierre Hardy
- Department of Pediatrics, Physiology and Pharmacology, Université de Montréal, H3C 3J7 Montréal, Québec, Canada
| | - Jeanne Leblond Chain
- Gene Delivery Laboratory, Faculty of Pharmacy, Université de Montréal, H3C 3J7 Montréal, Québec, Canada; Université de Bordeaux, ARNA Laboratory, INSERM U1212, CNRS UMR 5320, F-33016 Bordeaux, France.
| |
Collapse
|
4
|
Yoo B, Meka N, Sheedy P, Billig AM, Pantazopoulos P, Medarova Z. MicroRNA-710 regulates multiple pathways of carcinogenesis in murine metastatic breast cancer. PLoS One 2019; 14:e0226356. [PMID: 31834924 PMCID: PMC6910689 DOI: 10.1371/journal.pone.0226356] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/25/2019] [Indexed: 11/18/2022] Open
Abstract
Prior research has shown that critical differences between non-metastatic and metastatic tumor cells are at the level of microRNA. Consequently, harnessing these molecules for the treatment of metastatic cancer could have significant clinical impact. In the present study, we set out to identify metastasis-specific microRNAs which drive metastatic colonization of distant organs. Using a murine model of metastatic breast cancer, we employed a directed approach in which we screened for microRNAs that are differentially expressed between the primary tumors and metastatic lesions but concordantly expressed in all of the metastatic lesions irrespective of the tissue that is colonized. Of the identified targets, we focused on miR-710, which was consistently and significantly downregulated in the metastatic lesions relative to the primary tumors. The level of downregulation was independent of the distant organ that is involved, suggesting that miR-710 plays a fundamental role in metastatic colonization. Computational target prediction suggested a pleiotropic role for miR-710 in apoptosis, migration and invasion, and stemness. Using a previously validated oligonucleotide delivery system, we introduced miR-710 mimics into 4T1 metastatic breast adenocarcinoma cells and assessed the resultant phenotypic effects. We demonstrated significant inhibition of cell viability, migration, and invasion. We also showed that the treatment profoundly enhanced cell senescence, reduced stemness, and influenced markers of epithelial to mesenchymal transition, as evidenced by enhanced E-cadherin and reduced vimentin expression. This knowledge represents a first step towards harnessing a similar approach to discover novel microRNA targets with therapeutic potential in metastasis.
Collapse
Affiliation(s)
- Byunghee Yoo
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States of America
- * E-mail: (BY); (ZM)
| | - Nikhil Meka
- College of Arts and Science, New York University, NY, United States of America
| | - Patrick Sheedy
- Department of Health Sciences, CaNCURE Program, Northeastern University, Boston, MA
| | - Ann-Marie Billig
- Department of Health Sciences, CaNCURE Program, Northeastern University, Boston, MA
| | - Pamela Pantazopoulos
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Zdravka Medarova
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States of America
- * E-mail: (BY); (ZM)
| |
Collapse
|
5
|
Li F, Aljahdali I, Ling X. Cancer therapeutics using survivin BIRC5 as a target: what can we do after over two decades of study? JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:368. [PMID: 31439015 PMCID: PMC6704566 DOI: 10.1186/s13046-019-1362-1] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 08/06/2019] [Indexed: 02/06/2023]
Abstract
Survivin (also named BIRC5) is a well-known cancer therapeutic target. Since its discovery more than two decades ago, the use of survivin as a target for cancer therapeutics has remained a central goal of survivin studies in the cancer field. Many studies have provided intriguing insight into survivin's functional role in cancers, thus providing promise for survivin as a cancer therapeutic target. Despite this, moving survivin-targeting agents into and through the clinic remains a challenge. In order to address this challenge, we may need to rethink current strategies in order to develop a new mindset for targeting survivin. In this Review, we will first summarize the current survivin mechanistic studies, and then review the status of survivin cancer therapeutics, which is classified into five categories: (i) survivin-partner protein interaction inhibitors, (ii) survivin homodimerization inhibitors, (iii) survivin gene transcription inhibitors, (iv) survivin mRNA inhibitors and (v) survivin immunotherapy. We will then provide our opinions on cancer therapeutics using survivin as a target, with the goal of stimulating discussion that might facilitate translational research for discovering improved strategies and/or more effective anticancer agents that target survivin for cancer therapy.
Collapse
Affiliation(s)
- Fengzhi Li
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, New York, 14263, USA. .,Developmental Therapeutics Program, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, New York, 14263, USA.
| | - Ieman Aljahdali
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, New York, 14263, USA.,Department of Cellular & Molecular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, New York, 14263, USA
| | - Xiang Ling
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, New York, 14263, USA.,Canget BioTekpharma LLC, Buffalo, New York, USA
| |
Collapse
|
6
|
RNAi-Mediated PD-L1 Inhibition for Pancreatic Cancer Immunotherapy. Sci Rep 2019; 9:4712. [PMID: 30886310 PMCID: PMC6423142 DOI: 10.1038/s41598-019-41251-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 03/05/2019] [Indexed: 12/21/2022] Open
Abstract
The recent past has seen impressive progress in the treatment of various malignancies using immunotherapy. One of the most promising approaches involves immune checkpoint inhibitors. However, the clinical results with these agents have demonstrated variability in the response. Pancreatic cancer, in particular, has proven resistant to initial immunotherapy approaches. Here, we describe an alternative strategy that relies on combining gemcitabine and a novel programmed death-ligand 1 (PD-L1) inhibitor, termed MN-siPDL1. MN-siPDL1 incorporates small interfering RNA against PD-L1 (siPDL1) conjugated to a magnetic nanocarrier (MN). We show that noninvasive magnetic resonance imaging (MRI) could be used to monitor therapeutic response. Combination therapy consisting of gemcitabine and MN-siPDL1 in a syngeneic murine pancreatic cancer model resulted in a significant reduction in tumor growth and an increase in survival. Following optimization, a 90% reduction in tumor volume was achieved 2 weeks after the beginning of treatment. Whereas 100% of the control animals had succumbed to their tumors by week 6 after the beginning of treatment, there was no mortality in the experimental group by week 5, and 67% of the experimental animals survived for 12 weeks. This method could provide therapeutic benefit against an intractable disease for which there are no effective treatments and which is characterized by a mere 1% 5-year survival.
Collapse
|
7
|
Daglioglu C, Kaci FN. Cascade therapy with doxorubicin and survivin-targeted tailored nanoparticles: An effective alternative for sensitization of cancer cells to chemotherapy. Int J Pharm 2019; 561:74-81. [PMID: 30825555 DOI: 10.1016/j.ijpharm.2019.02.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/14/2019] [Accepted: 02/18/2019] [Indexed: 12/15/2022]
Abstract
Chemotherapy frequently involves combination treatment protocols to maximize tumor cell killing. Unfortunately these intensive chemotherapeutic regimes, often show disappointing results due to the development of drug resistance and higher nonspecific toxicity on normal tissues. In cancer treatment, it is critically important to minimize toxicity while preserving efficacy. We have previously addressed this issue and proposed a nanoparticle-based combination therapy involving both a molecularly targeted therapy and chemotherapeutic agent for neutralizing antiapoptotic survivin (BIRC5) to potentiate the efficacy of doxorubicin (DOX). Although the particles exhibited strong anticancer effect on the lung carcinoma A549 and the cervical carcinoma HeLa cells, there were lower-level therapeutic outcomes on the colon carcinoma HCT-116, the leukemia Jurkat and the pancreatic carcinoma MIA PaCa-2 cells. Since targeted therapies are one of the key approaches for overcoming drug resistance, tailoring the treatment of cancer cells with distinct characteristics is necessary to improve the therapeutic outcome of cancer therapy and to minimize potential pharmacokinetic interactions of drugs. In the light of this issue, this study examined whether a cascade therapy with low-dose DOX and survivin-targeted tailored nanoparticles is more effective at sensitizing HCT-116, Jurkat and MIA PaCa-2 cancer cells to DOX-chemotherapy than simultaneous combination therapy. The results demonstrated that the sequential therapy with the protocol comprising addition of the nanoparticles after incubation of cells with DOX clearly advanced the therapeutic outcome of related cancer cells, whereas the reverse protocol resulted in a reduction or delay in apoptosis, emphasizing the critical importance of formulating synergistic drug combinations in cancer therapy.
Collapse
Affiliation(s)
- Cenk Daglioglu
- Izmir Institute of Technology, Faculty of Science, Department of Molecular Biology and Genetics, Urla/Izmir 35430, Turkey.
| | - Fatma Necmiye Kaci
- Erzurum Technical University, Faculty of Science, Department of Molecular Biology and Genetics, Yakutiye/Erzurum 25050, Turkey
| |
Collapse
|
8
|
Liu X, Zhao Y, Zhang W, Gao Y, Huo M, Liu M, Xiao Z, Liang S, Xu N, Zhu H. Inhibition of survivin enhances radiosensitivity of esophageal cancer cells by switching radiation-induced senescence to apoptosis. Onco Targets Ther 2018; 11:3087-3100. [PMID: 29872320 PMCID: PMC5975611 DOI: 10.2147/ott.s166798] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Purpose Strategies to increase radiosensitivity are urgently needed. Combining radiosensitizing reagents with radiotherapy could improve the outcome of cancer treatment. Some preclinical studies showed that sepantronium bromide (YM155) could sensitize cancer cells to radiation by inhibiting the survivin protein. In this study, we try to investigate the function of YM155 on radiosensitivity of esophageal squamous cell carcinoma (ESCC) cells. Materials and methods ESCC cell lines were treated with radiation and YM155, and the radiation efficacy was evaluated by cell counting kit-8 assay and clonogenic survival assay. Cell senescence was measured by senescence-associated β-galactosidase staining. Terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling assay, fluorescein isothiocyanate-labeled Annexin V/propidium iodide assay, and poly ADP-ribose polymerase cleavage were used to detect apoptosis. KYSE150 xenografts model was used to test the efficacy of radiation combined with YM155. Results YM155 could inhibit the upregulation of survivin induced by radiation in all ESCC cell lines, but the efficacy of radiosensitization varied in different cell lines. Radiation-induced senescence in KYSE150 and KYSE410 cells, and the combination with YM155 inhibited senescence and promoted apoptosis of ESCC cells, thereby enhancing radiosensitivity. Combination with YM155 and radiation delayed the growth of KYSE150 xenografts in nude mice by switching radiation-induced senescence to apoptosis. When p21 was inhibited in KYSE150 cells, radiation did not induce senescence, and the radiosensitization of YM155 was also attenuated. In KYSE510 and KYSE180 cells, radiation did not induce senescence, and YM155 could not enhance the radiosensitivity. Conclusion Our results suggest a new mechanism that YM155 might sensitize ESCC cells to radiation by switching radiation-induced senescence to apoptosis. The major determinant of radiosensitization by YM155 might be the induction of senescence by radiation.
Collapse
Affiliation(s)
- Xianghe Liu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yahui Zhao
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weina Zhang
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yang Gao
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Miaomiao Huo
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mei Liu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zefen Xiao
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shufang Liang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Ningzhi Xu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Hongxia Zhu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
9
|
Cheng C, Wang T, Song Z, Peng L, Gao M, Hermine O, Rousseaux S, Khochbin S, Mi J, Wang J. Induction of autophagy and autophagy-dependent apoptosis in diffuse large B-cell lymphoma by a new antimalarial artemisinin derivative, SM1044. Cancer Med 2018; 7:380-396. [PMID: 29277967 PMCID: PMC5806110 DOI: 10.1002/cam4.1276] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 11/06/2017] [Accepted: 11/07/2017] [Indexed: 12/11/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common form of non-Hodgkin's lymphoma. R-CHOP is currently the standard therapy for DLBCL, but the prognosis of refractory or recurrent patients remains poor. In this study, we synthesized a new water-soluble antimalarial drug artemisinin derivative, SM1044. The treatment of DLBCL cell lines with SM1044 induces autophagy-dependent apoptosis, which is directed by an accelerated degradation of the antiapoptosis protein Survivin, via its acetylation-dependent interaction with the autophagy-related protein LC3-II. Additionally, SM1044 also stimulates the de novo synthesis of ceramide, which in turn activates the CaMKK2-AMPK-ULK1 axis, leading to the initiation of autophagy. Our findings not only elucidate the mechanism of autophagy-dependent apoptosis in DLBCL cells, but also suggest that SM1044 is a promising therapeutic molecule for the treatment of DLBCL, along with R-CHOP regimen.
Collapse
Affiliation(s)
- Chunyan Cheng
- State Key Laboratory for Medical GenomicsDepartment of HematologyShanghai Institute of HematologyCollaborative Innovation Center of Systems BiomedicinePôle Sino‐Français des Sciences du Vivant et GenomiqueRui Jin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Tao Wang
- State Key Laboratory for Medical GenomicsDepartment of HematologyShanghai Institute of HematologyCollaborative Innovation Center of Systems BiomedicinePôle Sino‐Français des Sciences du Vivant et GenomiqueRui Jin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhiqun Song
- Department of Blood Transfusionthe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuChina
| | - Lijun Peng
- State Key Laboratory for Medical GenomicsDepartment of HematologyShanghai Institute of HematologyCollaborative Innovation Center of Systems BiomedicinePôle Sino‐Français des Sciences du Vivant et GenomiqueRui Jin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Mengqing Gao
- State Key Laboratory for Medical GenomicsDepartment of HematologyShanghai Institute of HematologyCollaborative Innovation Center of Systems BiomedicinePôle Sino‐Français des Sciences du Vivant et GenomiqueRui Jin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Olivier Hermine
- Service d'Hématologie AdultesHôpital Necker‐Enfants MaladesAssistance Publique‐Hôpitaux de ParisUniversité Paris DescartesParisFrance
| | - Sophie Rousseaux
- CNRS UMR 5309/INSERM U1209/Université Grenoble‐Alpes/Institute for Advanced BiosciencesLa TroncheFrance
| | - Saadi Khochbin
- CNRS UMR 5309/INSERM U1209/Université Grenoble‐Alpes/Institute for Advanced BiosciencesLa TroncheFrance
| | - Jian‐Qing Mi
- State Key Laboratory for Medical GenomicsDepartment of HematologyShanghai Institute of HematologyCollaborative Innovation Center of Systems BiomedicinePôle Sino‐Français des Sciences du Vivant et GenomiqueRui Jin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jin Wang
- State Key Laboratory for Medical GenomicsDepartment of HematologyShanghai Institute of HematologyCollaborative Innovation Center of Systems BiomedicinePôle Sino‐Français des Sciences du Vivant et GenomiqueRui Jin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
10
|
Yoo B, Billig AM, Medarova Z. Guidelines for Rational Cancer Therapeutics. Front Oncol 2017; 7:310. [PMID: 29312885 PMCID: PMC5732930 DOI: 10.3389/fonc.2017.00310] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 11/29/2017] [Indexed: 12/12/2022] Open
Abstract
Traditionally, cancer therapy has relied on surgery, radiation therapy, and chemotherapy. In recent years, these interventions have become increasingly replaced or complemented by more targeted approaches that are informed by a deeper understanding of the underlying biology. Still, the implementation of fully rational patient-specific drug design appears to be years away. Here, we present a vision of rational drug design for cancer that is defined by two major components: modularity and image guidance. We suggest that modularity can be achieved by combining a nanocarrier and an oligonucleotide component into the therapeutic. Image guidance can be incorporated into the nanocarrier component by labeling with a specific imaging reporter, such as a radionuclide or contrast agent for magnetic resonance imaging. While limited by the need for additional technological advancement in the areas of cancer biology, nanotechnology, and imaging, this vision for the future of cancer therapy can be used as a guide to future research endeavors.
Collapse
Affiliation(s)
- Byunghee Yoo
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Ann-Marie Billig
- Bouvé College of Health Sciences, Northeastern University, Boston, MA, United States
| | - Zdravka Medarova
- MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
11
|
Sequential therapy with redox-responsive glucolipid nanocarrier separately delivering siRNA and doxorubicin to overcome multidrug resistance. Int J Pharm 2017; 534:368-377. [DOI: 10.1016/j.ijpharm.2017.10.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 09/22/2017] [Accepted: 10/15/2017] [Indexed: 12/12/2022]
|
12
|
Elnegaard MP, List M, Christiansen H, Schmidt S, Mollenhauer J, Block I. Protein-based nanotoxicology assessment strategy. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:1229-1233. [DOI: 10.1016/j.nano.2016.12.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/06/2016] [Accepted: 12/25/2016] [Indexed: 02/07/2023]
|
13
|
Lo ST, Pantazopouos P, Medarova Z, Moore A. Presentation of underglycosylated mucin 1 in pancreatic adenocarcinoma (PDAC) at early stages. Am J Cancer Res 2016; 6:1986-1995. [PMID: 27725904 PMCID: PMC5043108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 08/06/2016] [Indexed: 06/06/2023] Open
Abstract
Underglycosylated mucin 1 antigen (uMUC1) is a proven biomarker of cancer progression relevant to many malignancies including pancreatic ductal adenocarcinoma (PDAC). However, while ample evidence exists of the expression of total MUC1, little is known about the abundance of the underglycolsylated form of the antigen and its significance in disease progression. Such knowledge is important because the underglycosylated form of MUC1 is intimately linked to metastatic potential. Here, we investigated the expression uMUC1 at various stages of PDAC including pancreatic intraepithelial neoplasia (PanIN). Immunohistochemical analysis was performed on human tissue microarrays (TMAs) containing PDAC and PanIN using monoclonal antibody specific to uMUC1. uMUC1 expression was analyzed by a traditional pathological scoring system and using automatic imaging analysis software. Our results demonstrated low uMUC1 abundance in PanIN lesions and a transient increase in antigen availability in stage I PDAC, followed by decreased expression in later stages of the disease. An additional finding was that there was intermediate expression of uMUC1 in adjacent normal tissues from PDAC irrespective of the stage. These studies suggest the intriguing possibility that a pro-metastatic uMUC1 expression signature may appear at early stages of PDAC, providing an additional clue about the aggressive nature of pancreatic cancer.
Collapse
Affiliation(s)
- Su-Tang Lo
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical SchoolCharlestown, MA 02129, USA
- Current address: Advanced Imaging Research Center, UT Southwestern Medical CenterDallas, TX 75390, USA
| | - Pamela Pantazopouos
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical SchoolCharlestown, MA 02129, USA
| | - Zdravka Medarova
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical SchoolCharlestown, MA 02129, USA
| | - Anna Moore
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical SchoolCharlestown, MA 02129, USA
| |
Collapse
|
14
|
Daglioglu C, Okutucu B. Synthesis and Characterization of AICAR and DOX Conjugated Multifunctional Nanoparticles as a Platform for Synergistic Inhibition of Cancer Cell Growth. Bioconjug Chem 2016; 27:1098-111. [PMID: 26996194 DOI: 10.1021/acs.bioconjchem.6b00080] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The success of cancer treatment depends on the response to chemotherapeutic agents. However, malignancies often acquire resistance to drugs if they are used frequently. Combination therapy involving both a chemotherapeutic agent and molecularly targeted therapy may have the ability to retain and enhance therapeutic efficacy. Here, we addressed this issue by examining the efficacy of a novel therapeutic strategy that combines AICAR and DOX within a multifunctional platform. In this context, we reported the bottom-up synthesis of Fe3O4@SiO2(FITC)-FA/AICAR/DOX multifunctional nanoparticles aiming to neutralize survivin (BIRC5) to potentiate the efficacy of DOX against chemoresistance. The structure of nanoparticles was characterized by dynamic light scattering (DLS), zeta-potential measurement, X-ray diffraction (XRD), Fourier transform infrared spectroscopy (FT-IR), thermogravimetric analysis (TGA), and electron microscopy (SEM and STEM with EDX) techniques. Cellular uptake and cytotoxicity experiments demonstrated preferentially targeted delivery of nanoparticles and an efficient reduction of cancer cell viability in five different tumor-derived cell lines (A549, HCT-116, HeLa, Jurkat, and MIA PaCa-2). These results indicate that the multifunctional nanoparticle system possesses high inhibitory drug association and sustained cytotoxic effect with good biocompatibility. This novel approach which combines AICAR and DOX within a single platform might be promising as an antitumor treatment for cancer.
Collapse
Affiliation(s)
- Cenk Daglioglu
- Ege University , Faculty of Science, Biochemistry Department, Bornova/Izmir 35040, Turkey.,Izmir Institute of Technology, Faculty of Science, Department of Molecular Biology and Genetics, Urla/Izmir 35430, Turkey
| | - Burcu Okutucu
- Ege University , Faculty of Science, Biochemistry Department, Bornova/Izmir 35040, Turkey
| |
Collapse
|
15
|
Ma K, Xu Q, Wang S, Zhang W, Liu M, Liang S, Zhu H, Xu N. Nuclear accumulation of Yes-Associated Protein (YAP) maintains the survival of doxorubicin-induced senescent cells by promoting survivin expression. Cancer Lett 2016; 375:84-91. [PMID: 26944315 DOI: 10.1016/j.canlet.2016.02.045] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 02/23/2016] [Accepted: 02/24/2016] [Indexed: 02/05/2023]
Abstract
Although chemotherapeutic drugs can induce senescence to prohibit further division of tumor cells, senescence could also promote tumorigenesis mainly through a senescence-associated secretory phenotype. Therefore, senescent tumor cells should be eliminated immediately to prevent drug resistance and recurrence. Here, we used a doxorubicin-induced senescence model to explore the mechanism underlying the survival of therapy-induced senescent cells. After low-dose doxorubicin treatment, tumor cells turned on a senescence program and became large and flattened, increasing their contact area with the extracellular matrix (ECM). Furthermore, Yes-associated protein (YAP) accumulated in the nucleus and YAP activity was increased in doxorubicin-induced senescent cells. Knockdown of YAP increased the sensitivity of cells to low-dose doxorubicin treatment, causing apoptosis rather than senescence. Moreover, the anti-apoptotic gene survivin, a YAP target gene, was overexpressed in senescent cells. Inhibition of survivin could lead to selective elimination of senescent cells through apoptosis. Our study indicates that nuclear accumulation of YAP could promote the survival of senescent cells by increasing survivin expression. Therefore, targeting YAP or survivin might be a new strategy for clearing senescent cancer cells during drug treatment.
Collapse
Affiliation(s)
- Kai Ma
- Laboratory of Cell and Molecular Biology, State Key Laboratory of Molecular Oncology, Cancer Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Qing Xu
- Laboratory of Cell and Molecular Biology, State Key Laboratory of Molecular Oncology, Cancer Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Shuren Wang
- Laboratory of Cell and Molecular Biology, State Key Laboratory of Molecular Oncology, Cancer Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Weina Zhang
- Laboratory of Cell and Molecular Biology, State Key Laboratory of Molecular Oncology, Cancer Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Mei Liu
- Laboratory of Cell and Molecular Biology, State Key Laboratory of Molecular Oncology, Cancer Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Shufang Liang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, No.17, 3rd Section of People's South Road, Chengdu 610041, China
| | - Hongxia Zhu
- Laboratory of Cell and Molecular Biology, State Key Laboratory of Molecular Oncology, Cancer Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100021, China.
| | - Ningzhi Xu
- Laboratory of Cell and Molecular Biology, State Key Laboratory of Molecular Oncology, Cancer Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100021, China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, No.17, 3rd Section of People's South Road, Chengdu 610041, China.
| |
Collapse
|
16
|
Khan I, Agris PF, Yigit MV, Royzen M. In situ activation of a doxorubicin prodrug using imaging-capable nanoparticles. Chem Commun (Camb) 2016; 52:6174-7. [DOI: 10.1039/c6cc01024e] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A general strategy for image-guided prodrug activation using fluorescently-labeled magnetic iron oxide nanoparticles is described.
Collapse
Affiliation(s)
- Irfan Khan
- Department of Chemistry and The RNA Institute
- University at Albany
- State University of New York
- Albany
- USA
| | - Paul F. Agris
- Department of Chemistry and The RNA Institute
- University at Albany
- State University of New York
- Albany
- USA
| | - Mehmet V. Yigit
- Department of Chemistry and The RNA Institute
- University at Albany
- State University of New York
- Albany
- USA
| | - Maksim Royzen
- Department of Chemistry and The RNA Institute
- University at Albany
- State University of New York
- Albany
- USA
| |
Collapse
|
17
|
Controlling RNA Expression in Cancer Using Iron Oxide Nanoparticles Detectable by MRI and In Vivo Optical Imaging. Methods Mol Biol 2016; 1372:163-79. [PMID: 26530923 DOI: 10.1007/978-1-4939-3148-4_13] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Herein, we describe a protocol for the preparation of iron oxide nanoparticle-based contrast agents and drug delivery vehicles for noninvasive cancer imaging and therapy. In the first part of the chapter we describe the details of the contrast agent synthesis, functionalization, and characterization. In the second part we describe the methods for tumor imaging using the synthesized particles with noninvasive T2-weighted magnetic resonance imaging (MRI) and in vivo near infrared optical imaging.
Collapse
|
18
|
Jones SK, Lizzio V, Merkel OM. Folate Receptor Targeted Delivery of siRNA and Paclitaxel to Ovarian Cancer Cells via Folate Conjugated Triblock Copolymer to Overcome TLR4 Driven Chemotherapy Resistance. Biomacromolecules 2015; 17:76-87. [PMID: 26636884 DOI: 10.1021/acs.biomac.5b01189] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
This paper focuses on the ability of a folate-decorated triblock copolymer to deliver a targeted dose of siRNA in order to overcome chemotherapy resistance which can commonly cause complications in ovarian cancer patients. The micelleplexes that are formed upon electrostatic interaction with siRNA are used to deliver siRNA in a targeted manner to SKOV-3 ovarian cancer cells that overexpress folate receptor-α (FRα). The triblock copolymer consists of polyethylenimine-graft-polycaprolactone-block-poly(ethylene glycol) (PEI-g-PCL-b-PEG-Fol). In this work, polymers of different molecular weights of PEG, as well as different grafting degrees of the (g-PCL-b-PEG-Fol) chains to PEI, were analyzed to optimize targeted siRNA delivery. The polymers, their micelleplexes, and the in vitro performance of the latter were characterized by nuclear magnetic resonance, dynamic light scattering, transmission electron microscopy, flow cytrometry, western blot, confocal microscopy, and in luciferase assays. The different PEI-g-PCL-b-PEG-Fol conjugates showed suitable sizes below 260 nm, especially at N/P 5, which also allowed for full siRNA condensation. Furthermore, flow cytometry and Western blot analysis demonstrated that our best polymer was able to effectively deliver siRNA and that siRNA delivery resulted in efficient protein knockdown of toll-like receptor 4 (TLR4). Consequently, TLR4 knock down within SKOV-3 cells resensitized them toward paclitaxel (PTX) treatment, and apoptotic events increased. This study demonstrates that PEI-g-PCL-b-PEG-Fol conjugates are a reliable delivery system for siRNA and are able to mediate therapeutic protein knockdown within ovarian cancer cells. Additionally, this study provides further evidence to link TLR4 levels to chemotherapy resistance.
Collapse
Affiliation(s)
- Steven K Jones
- Department of Oncology, Wayne State University , 4100 John R Street, Detroit, Michigan 48201, United States
| | - Vincent Lizzio
- School of Medicine, Wayne State University , 540 East Canfield Street, Detroit, Michigan 48201, United States.,Department of Pharmaceutical Sciences, Wayne State University , 259 Mack Avenue, Detroit, Michigan 48201, United States
| | - Olivia M Merkel
- Department of Oncology, Wayne State University , 4100 John R Street, Detroit, Michigan 48201, United States.,Department of Pharmaceutical Sciences, Wayne State University , 259 Mack Avenue, Detroit, Michigan 48201, United States
| |
Collapse
|
19
|
Liu X, Wang J, Xu W, Ding J, Shi B, Huang K, Zhuang X, Chen X. Glutathione-degradable drug-loaded nanogel effectively and securely suppresses hepatoma in mouse model. Int J Nanomedicine 2015; 10:6587-602. [PMID: 26543363 PMCID: PMC4622485 DOI: 10.2147/ijn.s90000] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The reduction-responsive polymeric nanocarriers have attracted considerable interest because of a significantly higher concentration of intracellular glutathione in comparison with that outside cells. The smart nanovehicles can selectively transport the antitumor drugs into cells to improve efficacies and decrease side effects. In this work, a facilely prepared glutathione-degradable nanogel was employed for targeting intracellular delivery of an antitumor drug (ie, doxorubicin [DOX]). DOX was loaded into nanogel through a sequential dispersion and dialysis approach with a drug loading efficiency of 56.8 wt%, and the laden nanogel (noted as NG/DOX) showed an appropriate hydrodynamic radius of 56.1±3.5 nm. NG/DOX exhibited enhanced or improved maximum tolerated dose on healthy Kunming mice and enhanced intratumoral accumulation and dose-dependent antitumor efficacy toward H22 hepatoma-xenografted mouse model compared with free drug. In addition, the upregulated antitumor efficacy of NG/DOX was further confirmed by the histopathological and immunohistochemical analyses. Furthermore, the excellent in vivo security of NG/DOX was confirmed by the detection of body weight, histopathology, and biochemical indices of corresponding organs and serum. With controllable large-scale preparation and fascinating in vitro and in vivo properties, the reduction-responsive nanogel exhibited a good prospect for clinical chemotherapy.
Collapse
Affiliation(s)
- Xingang Liu
- Department of Critical Care Medicine, The First Hospital of Jilin University, Changchun, People’s Republic of China
| | - Jianmeng Wang
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, People’s Republic of China
| | - Weiguo Xu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Jilin University, Changchun, People’s Republic of China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Jilin University, Changchun, People’s Republic of China
| | - Bo Shi
- Center for Biological Experiment, College of Basic Medicine, Jilin University, Changchun, People’s Republic of China
| | - Kexin Huang
- Center for Biological Experiment, College of Basic Medicine, Jilin University, Changchun, People’s Republic of China
| | - Xiuli Zhuang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Jilin University, Changchun, People’s Republic of China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Jilin University, Changchun, People’s Republic of China
| |
Collapse
|
20
|
Yoo B, Kavishwar A, Ross A, Wang P, Tabassum DP, Polyak K, Barteneva N, Petkova V, Pantazopoulos P, Tena A, Moore A, Medarova Z. Combining miR-10b-Targeted Nanotherapy with Low-Dose Doxorubicin Elicits Durable Regressions of Metastatic Breast Cancer. Cancer Res 2015; 75:4407-15. [PMID: 26359455 PMCID: PMC4609288 DOI: 10.1158/0008-5472.can-15-0888] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 07/31/2015] [Indexed: 12/22/2022]
Abstract
The therapeutic promise of microRNA (miRNA) in cancer has yet to be realized. In this study, we identified and therapeutically exploited a new role for miR-10b at the metastatic site, which links its overexpression to tumor cell viability and proliferation. In the protocol developed, we combined a miR-10b-inhibitory nanodrug with low-dose anthracycline to achieve complete durable regressions of metastatic disease in a murine model of metastatic breast cancer. Mechanistic investigations suggested a potent antiproliferative, proapoptotic effect of the nanodrug in the metastatic cells, potentiated by a cell-cycle arrest produced by administration of the low-dose anthracycline. miR-10b was overexpressed specifically in cells with high metastatic potential, suggesting a role for this miRNA as a metastasis-specific therapeutic target. Taken together, our results implied the existence of pathways that regulate the viability and proliferation of tumor cells only after they have acquired the ability to grow at distant metastatic sites. As illustrated by miR-10b targeting, such metastasis-dependent apoptotic pathways would offer attractive targets for further therapeutic exploration.
Collapse
Affiliation(s)
- Byunghee Yoo
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Amol Kavishwar
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Alana Ross
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ping Wang
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Doris P Tabassum
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Natalia Barteneva
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Victoria Petkova
- Molecular Medicine Core, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Pamela Pantazopoulos
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Aseda Tena
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts
| | - Anna Moore
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Zdravka Medarova
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
21
|
ZHANG HONG, LI ZHIHONG, WANG KAIZHONG, REN PING. Combined treatment of XIAP-targeting shRNA and celecoxib synergistically inhibits the tumor growth of non-small cell lung cancer cells in vitro and in vivo. Oncol Rep 2014; 33:1079-88. [DOI: 10.3892/or.2014.3678] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 07/16/2014] [Indexed: 11/05/2022] Open
|
22
|
Zhou Y, Han G, Wang Y, Hu X, Li Z, Chen L, Bai W, Luo J, Zhang Y, Sun J, Yang X. Radiofrequency heat-enhanced chemotherapy for breast cancer: towards interventional molecular image-guided chemotherapy. Am J Cancer Res 2014; 4:1145-52. [PMID: 25250095 PMCID: PMC4165778 DOI: 10.7150/thno.10006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 08/08/2014] [Indexed: 12/20/2022] Open
Abstract
Breast cancer is the most common malignancy in women worldwide. Recent developments in minimally invasive interventional radiology techniques have significantly improved breast cancer treatment. This study aimed to develop a novel technique for the local management of breast cancers using radiofrequency heat (RFH). We performed both in vitro experiments using human breast cancer cells and in vivo validation in xenograft animal models with magnetic resonance imaging (MRI) and pathological correlation to investigate the feasibility of our approach. Four treatment groups, including (1) no treatment (control), (2) RFH-only, (3) chemo (doxorubicin)-only, and (4) combination therapy with both doxorubicin and RFH, were conducted in each experiment. In vitro combination therapy significantly decreased breast cancer cell proliferation while increased their apoptosis index compared to the other three groups. MRI demonstrated a significant tumor size reduction in animals treated with combination therapy compared to those receiving other treatments in vivo. Such result was further confirmed by pathological examination. In conclusion, our findings suggests that RFH can enhance the therapeutic efficiency of doxorubicin on breast cancers, thus establishing the basis for future development of interventional molecular image-guided local chemotherapy for breast malignancies.
Collapse
|