1
|
Leck LYW, Abd El-Aziz YS, McKelvey KJ, Park KC, Sahni S, Lane DJR, Skoda J, Jansson PJ. Cancer stem cells: Masters of all traits. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167549. [PMID: 39454969 DOI: 10.1016/j.bbadis.2024.167549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 10/01/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
Cancer is a heterogeneous disease, which contributes to its rapid progression and therapeutic failure. Besides interpatient tumor heterogeneity, tumors within a single patient can present with a heterogeneous mix of genetically and phenotypically distinct subclones. These unique subclones can significantly impact the traits of cancer. With the plasticity that intratumoral heterogeneity provides, cancers can easily adapt to changes in their microenvironment and therapeutic exposure. Indeed, tumor cells dynamically shift between a more differentiated, rapidly proliferating state with limited tumorigenic potential and a cancer stem cell (CSC)-like state that resembles undifferentiated cellular precursors and is associated with high tumorigenicity. In this context, CSCs are functionally located at the apex of the tumor hierarchy, contributing to the initiation, maintenance, and progression of tumors, as they also represent the subpopulation of tumor cells most resistant to conventional anti-cancer therapies. Although the CSC model is well established, it is constantly evolving and being reshaped by advancing knowledge on the roles of CSCs in different cancer types. Here, we review the current evidence of how CSCs play a pivotal role in providing the many traits of aggressive tumors while simultaneously evading immunosurveillance and anti-cancer therapy in several cancer types. We discuss the key traits and characteristics of CSCs to provide updated insights into CSC biology and highlight its implications for therapeutic development and improved treatment of aggressive cancers.
Collapse
Affiliation(s)
- Lionel Y W Leck
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Yomna S Abd El-Aziz
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Oral Pathology Department, Faculty of Dentistry, Tanta University, Tanta, Egypt
| | - Kelly J McKelvey
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia
| | - Kyung Chan Park
- Proteina Co., Ltd./Seoul National University, Seoul, South Korea
| | - Sumit Sahni
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia
| | - Darius J R Lane
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience & Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Jan Skoda
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.
| | - Patric J Jansson
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
2
|
Logghe T, van Zwol E, Immordino B, Van den Cruys K, Peeters M, Giovannetti E, Bogers J. Hyperthermia in Combination with Emerging Targeted and Immunotherapies as a New Approach in Cancer Treatment. Cancers (Basel) 2024; 16:505. [PMID: 38339258 PMCID: PMC10854776 DOI: 10.3390/cancers16030505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/10/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
Despite significant advancements in the development of novel therapies, cancer continues to stand as a prominent global cause of death. In many cases, the cornerstone of standard-of-care therapy consists of chemotherapy (CT), radiotherapy (RT), or a combination of both. Notably, hyperthermia (HT), which has been in clinical use in the last four decades, has proven to enhance the effectiveness of CT and RT, owing to its recognized potency as a sensitizer. Furthermore, HT exerts effects on all steps of the cancer-immunity cycle and exerts a significant impact on key oncogenic pathways. Most recently, there has been a noticeable expansion of cancer research related to treatment options involving immunotherapy (IT) and targeted therapy (TT), a trend also visible in the research and development pipelines of pharmaceutical companies. However, the potential results arising from the combination of these innovative therapeutic approaches with HT remain largely unexplored. Therefore, this review aims to explore the oncology pipelines of major pharmaceutical companies, with the primary objective of identifying the principal targets of forthcoming therapies that have the potential to be advantageous for patients by specifically targeting molecular pathways involved in HT. The ultimate goal of this review is to pave the way for future research initiatives and clinical trials that harness the synergy between emerging IT and TT medications when used in conjunction with HT.
Collapse
Affiliation(s)
- Tine Logghe
- Elmedix NV, Dellingstraat 34/1, 2800 Mechelen, Belgium
| | - Eke van Zwol
- Elmedix NV, Dellingstraat 34/1, 2800 Mechelen, Belgium
| | - Benoît Immordino
- Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, San Giuliano, 56017 Pisa, Italy
- Institute of Life Sciences, Sant’Anna School of Advanced Studies, 56127 Pisa, Italy
| | | | - Marc Peeters
- Department of Oncology, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Elisa Giovannetti
- Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, San Giuliano, 56017 Pisa, Italy
- Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Johannes Bogers
- Elmedix NV, Dellingstraat 34/1, 2800 Mechelen, Belgium
- Laboratory of Cell Biology and Histology, Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| |
Collapse
|
3
|
Joshi G, Basu A. Epigenetic control of cell signalling in cancer stem cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 383:67-88. [PMID: 38359971 DOI: 10.1016/bs.ircmb.2023.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
The self-renewing cancer stem cells (CSCs) represent one of the distinct cell populations occurring in a tumour that can differentiate into multiple lineages. This group of sparsely abundant cells play a vital role in tumour survival and resistance to different treatments during cancer. The lack of exclusive markers associated with CSCs makes diagnosis and prognosis in cancer patients extremely difficult. This calls for the identification of unique regulators and markers for CSCs. Various signalling pathways like the Wnt/β-catenin pathway, Hedgehog pathway, Notch pathway, and TGFβ/BMP play a major role in the regulation and maintenance of CSCs. Epigenetic regulatory mechanisms add another layer of complexity to control these signalling pathways. In this chapter, we discuss about the role of epigenetic mechanisms in regulating the cellular signalling pathways in CSCs. The epigenetic regulatory mechanisms such as DNA methylation, histone modification and microRNAs can modulate the diverse effectors of signalling pathways and consequently the growth, differentiation and tumorigenicity of CSCs. In the end, we briefly discuss the therapeutic potential of targeting these epigenetic regulators and their target genes in CSCs.
Collapse
Affiliation(s)
- Gaurav Joshi
- Institute of Molecular Biology (IMB), Mainz, Germany.
| | - Amitava Basu
- Institute of Molecular Biology (IMB), Mainz, Germany.
| |
Collapse
|
4
|
Liu Z, Zhang C, Cui B, Wang Y, Lim K, Li K, Thiery JP, Chen J, Ho CL. Targeted EpCAM-binding for the development of potent and effective anticancer proteins. Biomed Pharmacother 2023; 161:114443. [PMID: 36863098 DOI: 10.1016/j.biopha.2023.114443] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/13/2023] [Accepted: 02/22/2023] [Indexed: 03/04/2023] Open
Abstract
Protein-based cancer therapies are considered an alternative to conventional anticancer regimens, providing multifunctional properties while showing low toxicity. However, its widespread use is limited by absorption and instability issues, resulting in higher dosage requirements and a prolonged onset of bioactivity to elicit the desired response. Here, we developed a non-invasive antitumor treatment using designed ankyrin repeat protein (DARPin)-anticancer protein-conjugate that specifically targets the cancer biomarker, epithelial cell adhesion molecule (EpCAM). The DARPin-anticancer proteins bind to EpCAM-positive cancer cells and improve the in vitro anticancer efficacy by over 100-folds within 24 h, where the DARPin-tagged human lactoferrin fragment (drtHLF4) IC50 value is within the nanomolar range. Orally administered drtHLF4 was readily absorbed into the systemic flow of the HT-29 cancer murine model, exerting its anticancer effect on other tumors in the host body. Orally administered drtHFL4 cleared HT29-colorectal tumors using a single dose, whereas intratumoral injection cleared HT29-subcutaneous tumors within three doses. This approach addresses the limitations of other protein-based anticancer treatments by providing a non-invasive anticancer therapy with improved potency and tumor-specificity.
Collapse
Affiliation(s)
- Zhao Liu
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, China.
| | - Chen Zhang
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, China.
| | - Beiming Cui
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, China; Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Yijie Wang
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, China.
| | - Kaisheng Lim
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, China; Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Kai Li
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, China.
| | - Jean Paul Thiery
- Guangzhou Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong Province, China.
| | - Jun Chen
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, China; Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Chun Loong Ho
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, China; Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|
5
|
Haque MR, Wessel CR, Leary DD, Wang C, Bhushan A, Bishehsari F. Patient-derived pancreatic cancer-on-a-chip recapitulates the tumor microenvironment. MICROSYSTEMS & NANOENGINEERING 2022; 8:36. [PMID: 35450328 PMCID: PMC8971446 DOI: 10.1038/s41378-022-00370-6] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/19/2022] [Accepted: 02/20/2022] [Indexed: 05/08/2023]
Abstract
The patient population suffering from pancreatic ductal adenocarcinoma (PDAC) presents, as a whole, with a high degree of molecular tumor heterogeneity. The heterogeneity of PDAC tumor composition has complicated treatment and stalled success in clinical trials. Current in vitro techniques insufficiently replicate the intricate stromal components of PDAC tumor microenvironments (TMEs) and fail to model a given tumor's unique genetic phenotype. The development of patient-derived organoids (PDOs) has opened the door for improved personalized medicine since PDOs are derived directly from patient tumors, thus preserving the tumors' unique behaviors and genetic phenotypes. This study developed a tumor-chip device engineered to mimic the PDAC TME by incorporating PDOs and stromal cells, specifically pancreatic stellate cells and macrophages. Establishing PDOs in a multicellular microfluidic chip device prolongs cellular function and longevity and successfully establishes a complex organotypic tumor environment that incorporates desmoplastic stroma and immune cells. When primary cancer cells in monoculture were subjected to stroma-depleting agents, there was no effect on cancer cell viability. However, targeting stroma in our tumor-chip model resulted in a significant increase in the chemotherapy effect on cancer cells, thus validating the use of this tumor-chip device for drug testing.
Collapse
Affiliation(s)
- Muhammad R. Haque
- Division of Digestive Diseases, Rush Center for Integrated Microbiome & Chronobiology Research, Rush University Medical Center, Chicago, IL 60612 USA
| | - Caitlin R. Wessel
- University of Louisville School of Medicine, Louisville, KY 40202 USA
| | - Daniel D. Leary
- Division of Digestive Diseases, Rush Center for Integrated Microbiome & Chronobiology Research, Rush University Medical Center, Chicago, IL 60612 USA
| | - Chengyao Wang
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL 60616 USA
| | - Abhinav Bhushan
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL 60616 USA
| | - Faraz Bishehsari
- Division of Digestive Diseases, Rush Center for Integrated Microbiome & Chronobiology Research, Rush University Medical Center, Chicago, IL 60612 USA
| |
Collapse
|
6
|
Haque MR, Barlass U, Armstrong A, Shaikh M, Bishehsari F. Novel role of the Mu-opioid receptor in pancreatic cancer: potential link between opioid use and cancer progression. Mol Cell Biochem 2022; 477:1339-1345. [PMID: 35138511 DOI: 10.1007/s11010-022-04377-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 01/27/2022] [Indexed: 10/19/2022]
Abstract
Opioids are the most popular drugs for both acute and chronic pain management. The G protein-coupled mu-opioid receptor (MOR) is the therapeutic target for most clinically used opioids, including morphine. A mounting number of publications suggest a relationship between the MOR and possible cancer progression and recurrence extending to managing chronic cancer pain. In this study, we studied the possible link between opioid use and pancreatic cancer (PC) progression. We found increased MOR expression in murine and human PC cell lines, human PC-derived organoids, and in the undifferentiated or poorly differentiated areas of surgically resected PC tissues. Direct stimulation of MOR by morphine (MOR agonist) caused a significant dose-dependent increase in proliferation, invasion, and levels of stemness markers in PC cells. In a co-culture system, MOR stimulation of macrophages also resulted in increased proliferation of PC cells. MOR overexpression increased proliferation and cancer stemness, whereas knock-down of MOR followed opposite results in the PC cells. Morphine induced chemoresistance to conventional chemotherapeutic agents used for PC treatment. Overall, our results suggest that MOR is expressed in pancreatic cancer and may be involved in tumor progression and chemoresistance.
Collapse
Affiliation(s)
- Muhammad R Haque
- Division of Digestive Diseases, Rush Center for Integrated Microbiome & Chronobiology Research, Rush University Medical Center, 1725 W Harrison St, Chicago, IL, 60612, USA
| | - Usman Barlass
- Division of Digestive Diseases, Rush Center for Integrated Microbiome & Chronobiology Research, Rush University Medical Center, 1725 W Harrison St, Chicago, IL, 60612, USA
| | - Andrew Armstrong
- Division of Digestive Diseases, Rush Center for Integrated Microbiome & Chronobiology Research, Rush University Medical Center, 1725 W Harrison St, Chicago, IL, 60612, USA
| | - Maliha Shaikh
- Division of Digestive Diseases, Rush Center for Integrated Microbiome & Chronobiology Research, Rush University Medical Center, 1725 W Harrison St, Chicago, IL, 60612, USA
| | - Faraz Bishehsari
- Division of Digestive Diseases, Rush Center for Integrated Microbiome & Chronobiology Research, Rush University Medical Center, 1725 W Harrison St, Chicago, IL, 60612, USA.
| |
Collapse
|
7
|
Wu Y, Yi M, Zhu S, Wang H, Wu K. Recent advances and challenges of bispecific antibodies in solid tumors. Exp Hematol Oncol 2021; 10:56. [PMID: 34922633 PMCID: PMC8684149 DOI: 10.1186/s40164-021-00250-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/03/2021] [Indexed: 12/29/2022] Open
Abstract
Cancer immunotherapy has made remarkable progress in the past decade. Bispecific antibodies (BsAbs) have acquired much attention as the next generation strategy of antibody-target cancer immunotherapy, which overwhelmingly focus on T cell recruitment and dual receptors blockade. So far, BsAb drugs have been proved clinically effective and approved for the treatment of hematologic malignancies, but no BsAb have been approved in solid tumors. Numerous designed BsAb drugs for solid tumors are now undergoing evaluation in clinical trials. In this review, we will introduce the formats of bispecific antibodies, and then update the latest preclinical studies and clinical trials in solid tumors of BsAbs targeting EpCAM, CEA, PMSA, ErbB family, and so on. Finally, we discuss the BsAb-related adverse effects and the alternative strategy for future study.
Collapse
Affiliation(s)
- Yuze Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ming Yi
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shuangli Zhu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Haiyong Wang
- Beijing Anjianxi Medicinal Technology Co., Ltd., No.2 Cuiwei Road, Haidian District, Beijing, 100036, China.
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
8
|
Cao J, Bhatnagar S, Wang J, Qi X, Prabha S, Panyam J. Cancer stem cells and strategies for targeted drug delivery. Drug Deliv Transl Res 2021; 11:1779-1805. [PMID: 33095384 PMCID: PMC8062588 DOI: 10.1007/s13346-020-00863-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2020] [Indexed: 12/23/2022]
Abstract
Cancer stem cells (CSCs) are a small proportion of cancer cells with high tumorigenic activity, self-renewal ability, and multilineage differentiation potential. Standard anti-tumor therapies including conventional chemotherapy, radiation therapy, and molecularly targeted therapies are not effective against CSCs, and often lead to enrichment of CSCs that can result in tumor relapse. Therefore, it is hypothesized that targeting CSCs is key to increasing the efficacy of cancer therapies. In this review, CSC properties including CSC markers, their role in tumor growth, invasiveness, metastasis, and drug resistance, as well as CSC microenvironment are discussed. Further, CSC-targeted strategies including the use of targeted drug delivery systems are examined.
Collapse
Affiliation(s)
- Jin Cao
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
- College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Shubhmita Bhatnagar
- College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
- School of Pharmacy, Temple University, Philadelphia, PA, 19140, USA
| | - Jiawei Wang
- College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
- College of Pharmacy, University of Texas at Austin, Austin, TX, 78712, USA
| | - Xueyong Qi
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Swayam Prabha
- College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
- Cancer Research & Molecular Biology and Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Jayanth Panyam
- College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA.
- School of Pharmacy, Temple University, Philadelphia, PA, 19140, USA.
| |
Collapse
|
9
|
Herr I, Groth A, Salnikov AV, Büchler MW, Moldenhauer G. Combining forces to hit cancer stem cells: TRAIL-lymphocytes and EpCAMxCD3 bispecific antibody show efficacy against pancreatic cancer. Oncoimmunology 2021; 1:741-743. [PMID: 22934267 PMCID: PMC3429579 DOI: 10.4161/onci.19532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
TRAIL selectively kills cancer cells while bispecific antibody EpCAMxCD3 guides effector lymphocytes to cancer cells. Arming of ex vivo constructed TRAIL-lymphocytes with EpCAMxCD3 enhances contact time and affinity between lymphocytes and tumor cells and enforces tumor elimination. This boosts endogenous immune responses and augments the effect of cytotoxic tumor therapy.
Collapse
Affiliation(s)
- Ingrid Herr
- Molecular OncoSurgery Group; University of Heidelberg and German Cancer Research Center; Heidelberg, Germany
| | | | | | | | | |
Collapse
|
10
|
Yang H, Liu R, Xu Y, Qian L, Dai Z. Photosensitizer Nanoparticles Boost Photodynamic Therapy for Pancreatic Cancer Treatment. NANO-MICRO LETTERS 2021; 13:35. [PMID: 34138222 PMCID: PMC8187547 DOI: 10.1007/s40820-020-00561-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 10/31/2020] [Indexed: 05/13/2023]
Abstract
Patients with pancreatic cancer (PCa) have a poor prognosis apart from the few suitable for surgery. Photodynamic therapy (PDT) is a minimally invasive treatment modality whose efficacy and safety in treating unresectable localized PCa have been corroborated in clinic. Yet, it suffers from certain limitations during clinical exploitation, including insufficient photosensitizers (PSs) delivery, tumor-oxygenation dependency, and treatment escape of aggressive tumors. To overcome these obstacles, an increasing number of researchers are currently on a quest to develop photosensitizer nanoparticles (NPs) by the use of a variety of nanocarrier systems to improve cellular uptake and biodistribution of photosensitizers. Encapsulation of PSs with NPs endows them significantly higher accumulation within PCa tumors due to the increased solubility and stability in blood circulation. A number of approaches have been explored to produce NPs co-delivering multi-agents affording PDT-based synergistic therapies for improved response rates and durability of response after treatment. This review provides an overview of available data regarding the design, methodology, and oncological outcome of the innovative NPs-based PDT of PCa.
Collapse
Affiliation(s)
- Huanyu Yang
- Department of Ultrasound, Beijing Friendship Hospital, Capital Medical University, No. 95 Yongan Road, Xicheng District, Beijing, 100050, People's Republic of China
| | - Renfa Liu
- Department of Biomedical Engineering, College of Engineering, Peking University, No. 5 Yiheyuan Road, Haidian District, Beijing, 100871, People's Republic of China
| | - Yunxue Xu
- Department of Biomedical Engineering, College of Engineering, Peking University, No. 5 Yiheyuan Road, Haidian District, Beijing, 100871, People's Republic of China
| | - Linxue Qian
- Department of Ultrasound, Beijing Friendship Hospital, Capital Medical University, No. 95 Yongan Road, Xicheng District, Beijing, 100050, People's Republic of China.
| | - Zhifei Dai
- Department of Biomedical Engineering, College of Engineering, Peking University, No. 5 Yiheyuan Road, Haidian District, Beijing, 100871, People's Republic of China.
| |
Collapse
|
11
|
Selvanesan BC, Meena K, Beck A, Meheus L, Lara O, Rooman I, Gravekamp C. Nicotinamide combined with gemcitabine is an immunomodulatory therapy that restrains pancreatic cancer in mice. J Immunother Cancer 2020; 8:e001250. [PMID: 33154149 PMCID: PMC7646363 DOI: 10.1136/jitc-2020-001250] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Treatments for pancreatic ductal adenocarcinoma are poorly effective, at least partly due to the tumor's immune-suppressive stromal compartment. New evidence of positive effects on immune responses in the tumor microenvironment (TME), compelled us to test the combination of gemcitabine (GEM), a standard chemotherapeutic for pancreatic cancer, with nicotinamide (NAM), the amide form of niacin (vitamin B3), in mice with pancreatic cancer. METHODS Various mouse tumor models of pancreatic cancer, that is, orthotopic Panc-02 and KPC (KrasG12D, p53R172H, Pdx1-Cre) grafts, were treated alternately with NAM and GEM for 2 weeks, and the effects on efficacy, survival, stromal architecture and tumor-infiltrating immune cells was examined by immunohistochemistry (IHC), flow cytometry, Enzyme-linked immunospot (ELISPOT), T cell depletions in vivo, Nanostring analysis and RNAscope. RESULTS A significant reduction in tumor weight and number of metastases was found, as well as a significant improved survival of the NAM+GEM group compared with all control groups. IHC and flow cytometry showed a significant decrease in tumor-associated macrophages and myeloid-derived suppressor cells in the tumors of NAM+GEM-treated mice. This correlated with a significant increase in the number of CD4 and CD8 T cells of NAM+GEM-treated tumors, and CD4 and CD8 T cell responses to tumor-associated antigen survivin, most likely through epitope spreading. In vivo depletions of T cells demonstrated the involvement of CD4 T cells in the eradication of the tumor by NAM+GEM treatment. In addition, remodeling of the tumor stroma was observed with decreased collagen I and lower expression of hyaluronic acid binding protein, reorganization of the immune cells into lymph node like structures and CD31 positive vessels. Expression profiling for a panel of immuno-oncology genes revealed significant changes in genes involved in migration and activation of T cells, attraction of dendritic cells and epitope spreading. CONCLUSION This study highlights the potential of NAM+GEM as immunotherapy for advanced pancreatic cancer.
Collapse
Affiliation(s)
| | - Kiran Meena
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Amanda Beck
- Michael F. Price Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Lydie Meheus
- AntiCancer Fund, Boechoutlaan, Strombeek-Bever, Belgium
| | - Olaya Lara
- Laboratory of Medical and Molecular Oncology, Vrije Universiteit Brussel, Laarbeeklaan, Brussels, Belgium
| | - Ilse Rooman
- AntiCancer Fund, Boechoutlaan, Strombeek-Bever, Belgium
- Laboratory of Medical and Molecular Oncology, Vrije Universiteit Brussel, Laarbeeklaan, Brussels, Belgium
| | - Claudia Gravekamp
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
12
|
Hu Y, Wu Q, Gao J, Zhang Y, Wang Y. A meta-analysis and The Cancer Genome Atlas data of prostate cancer risk and prognosis using epithelial cell adhesion molecule (EpCAM) expression. BMC Urol 2019; 19:67. [PMID: 31324239 PMCID: PMC6642570 DOI: 10.1186/s12894-019-0499-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/09/2019] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Epithelial cell adhesion molecule (EpCAM) expression has been reported in many types of cancer, including prostate cancer (PCa). However, the role of EpCAM expression remains inconsistent. We conducted a meta-analysis to assess the clinicopathological and prognostic significance of EpCAM expression in PCa. METHODS Publications were searched online using electronic databases. The available data were obtained from The Cancer Genome Atlas (TCGA). The odds ratios (ORs) or hazard ratios (HRs) with their 95% confidence intervals (CIs) were calculated. RESULTS We identified seven studies in which immunohistochemistry was used and that included 871 prostatic tissue samples. EpCAM expression was significantly higher in PCa samples than in benign and normal tissue samples (OR = 77.93, P = 0.002; OR = 161.61, P < 0.001; respectively). No correlation of EpCAM overexpression with pT stage and lymph node metastasis was observed; however, EpCAM overexpression showed a significant correlation with Gleason score (OR = 0.48, P = 0.012) and bone metastasis (OR = 145.80, P < 0.001). Furthermore, TCGA data showed that EpCAM overexpression was not closely correlated with age, pT stage, lymph node metastasis, number of lymph node, prostate-specific antigen level, Gleason score, biochemical recurrence, and overall survival. Based on multivariate Cox proportional-hazards regression analysis, a significant correlation was observed between EpCAM overexpression and 5-year worse biochemical recurrence free-survival. CONCLUSIONS EpCAM overexpression may be correlated with the development of bone metastasis and worse biochemical recurrence free-survival of PCa. Further studies are needed to verify these findings.
Collapse
Affiliation(s)
- Yu Hu
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China
| | - Qiong Wu
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China
| | - Jialin Gao
- Department of Urology, The First Hospital of Jilin University, No. 71 Xinmin Street, Changchun, 130021, Jilin, China
| | - Yongrui Zhang
- Department of Urology, The First Hospital of Jilin University, No. 71 Xinmin Street, Changchun, 130021, Jilin, China
| | - Yuantao Wang
- Department of Urology, The First Hospital of Jilin University, No. 71 Xinmin Street, Changchun, 130021, Jilin, China.
| |
Collapse
|
13
|
Ni J, Cozzi P, Beretov J, Duan W, Bucci J, Graham P, Li Y. Epithelial cell adhesion molecule (EpCAM) is involved in prostate cancer chemotherapy/radiotherapy response in vivo. BMC Cancer 2018; 18:1092. [PMID: 30419852 PMCID: PMC6233586 DOI: 10.1186/s12885-018-5010-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 10/29/2018] [Indexed: 12/11/2022] Open
Abstract
Background Development of chemo−/radioresistance is a major challenge for the current prostate cancer (CaP) therapy. We have previously demonstrated that epithelial cell adhesion molecule (EpCAM) is associated with CaP growth and therapeutic resistance in vitro, however, the role of EpCAM in CaP in vivo is not fully elucidated. Here, we aimed to investigate how expression of EpCAM is involved in CaP growth and chemo−/radiotherapy response in NOD/SCID mouse models in vivo and to validate its role as a therapeutic target for CaP therapy. Methods EpCAM was knocked down in PC-3 CaP cell line using short hairpin RNA (shRNA). The effect of EpCAM-knockdown (KD) on tumour growth, chemo−/radiotherapy response and animal survival was evaluated on subcutaneous (s.c) and orthotopic mouse models. Results We found that KD of EpCAM significantly inhibited tumour growth, increased xenograft sensitivity to chemotherapy/radiotherapy, and prolonged the survival of tumour-bearing mice. In addition, we demonstrated that KD of EpCAM is associated with downregulation of the PI3K/Akt/mTOR pathway. Conclusions In conclusion, our data confirms that CaP growth and chemo−/radioresistance in vivo is associated with over-expression of EpCAM, which serves both a functional biomarker and promising therapeutic target. Electronic supplementary material The online version of this article (10.1186/s12885-018-5010-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jie Ni
- Cancer Care Centre, St George Hospital, Level 2, 4-10 South St, Kogarah, NSW, 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia
| | - Paul Cozzi
- St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia.,Department of Surgery, St George Hospital, Kogarah, NSW, 2217, Australia
| | - Julia Beretov
- Cancer Care Centre, St George Hospital, Level 2, 4-10 South St, Kogarah, NSW, 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia.,Anatomical Pathology, NSW Health Pathology, St George Hospital, Gray St, Kogarah, NSW, 2217, Australia
| | - Wei Duan
- School of Medicine and Centre for Molecular and Medical Research, Deakin University, Waurn Ponds, VIC, 3216, Australia
| | - Joseph Bucci
- Cancer Care Centre, St George Hospital, Level 2, 4-10 South St, Kogarah, NSW, 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia
| | - Peter Graham
- Cancer Care Centre, St George Hospital, Level 2, 4-10 South St, Kogarah, NSW, 2217, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia
| | - Yong Li
- Cancer Care Centre, St George Hospital, Level 2, 4-10 South St, Kogarah, NSW, 2217, Australia. .,St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia. .,School of Basic Medical Sciences, Zhengzhou University, Henan, 450001, China.
| |
Collapse
|
14
|
Dynamism, Sensitivity, and Consequences of Mesenchymal and Stem-Like Phenotype of Cancer Cells. Stem Cells Int 2018; 2018:4516454. [PMID: 30405720 PMCID: PMC6199882 DOI: 10.1155/2018/4516454] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 08/17/2018] [Indexed: 12/16/2022] Open
Abstract
There are remarkable similarities in the description of cancer stem cells (CSCs) and cancer cells with mesenchymal phenotype. Both cell types are highly tumorigenic, resistant against common anticancer treatment, and thought to cause metastatic growth. Moreover, cancer cells are able to switch between CSC and non-CSC phenotypes and vice versa, to ensure the necessary balance within the tumor. Likewise, cancer cells can switch between epithelial and mesenchymal phenotypes via well-described transition (EMT/MET) that is thought to be crucial for tumor propagation. In this review, we discuss whether, and to which extend, the CSCs and mesenchymal cancer cells are overlapping phenomena in terms of mechanisms, origin, and implication for cancer treatment. As well, we describe the dynamism of both phenotypes and involvement of the tumor microenvironment in CSC reversion and in EMT.
Collapse
|
15
|
Abstract
OPINION STATEMENT Managing patients with metastatic pancreatic adenocarcinoma (mPDA) is a challenging proposition for any treating oncologist. Although the potency of first-line therapies has improved with the approvals of FOLFIRINOX and gemcitabine plus nab-paclitaxel, many patients are unable to derive significant benefit from later lines of therapy upon progression. Enrollment on clinical trials remains among the best options for patients with mPDA in all lines of therapy. At our institution, we routinely check for microsatellite instability (MSI-H) and perform next-generation sequencing (NGS) at the time of diagnosis in all good performance status mPDA patients. Although MSI-H status is only found in 1% of patients with mPDA, given pembrolizumab's tissue-agnostic approval for MSI-H tumors in later-line settings, it is a viable option when deciding on subsequent lines of therapy. Any use of immune therapy in mPDA is investigational outside the MSI-H setting. NGS can identify BRCA or other DNA damage response (DDR) defects in patients which can predict sensitivity to platinum-based therapies and influence choice of both initial and later lines of therapy. It can also identify rare actionable genomic alterations such as HER2 (2%) and TRK fusions (0.1%) and offer patients the option of enrollment on clinical trials with agents targeting these or other identified alterations. We believe enrolling mPDA patients on clinical trials with immune-modulating agents is critical to determine if there are other patient subsets, outside of the MSI-H setting, who would benefit from these approaches. Immunotherapy's general tolerability and potential to generate durable responses make it particularly appealing for mPDA patients. Although single-modality immunotherapy such as checkpoint inhibitors or vaccines have not demonstrated efficacy in this disease, combinatorial strategies targeting unique aspects of PDA including the tumor microenvironment and desmoplastic stroma have shown preclinical or early-phase success. Validating these treatments with later-phase prospective studies is essential to making immunotherapy a routine component of the treatment armamentarium for mPDA patients.
Collapse
Affiliation(s)
- Satya Das
- Division of Hematology and Oncology, Department of Internal Medicine, Vanderbilt University Medical Center, 2220 Pierce Avenue, 777 Preston Research Building, Nashville, TN, 37232, USA.
| | - Jordan Berlin
- Division of Hematology and Oncology, Department of Internal Medicine, Vanderbilt University Medical Center, 2220 Pierce Avenue, 777 Preston Research Building, Nashville, TN, 37232, USA
| | - Dana Cardin
- Division of Hematology and Oncology, Department of Internal Medicine, Vanderbilt University Medical Center, 2220 Pierce Avenue, 777 Preston Research Building, Nashville, TN, 37232, USA
| |
Collapse
|
16
|
Abstract
Harnessing the power of the human immune system to treat cancer is the essence of immunotherapy. Monoclonal antibodies engage the innate immune system to destroy targeted cells. For the last 30years, antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity have been the main mechanisms of anti-tumor action of unconjugated antibody drugs. Efforts to exploit the potentials of other immune cells, in particular T cells, culminated in the recent approval of two T cell engaging bispecific antibody (T-BsAb) drugs, thereby stimulating new efforts to accelerate similar platforms through preclinical and clinical trials. In this review, we have compiled the worldwide effort in exploring T cell engaging bispecific antibodies. Our special emphasis is on the lessons learned, with the hope to derive insights in this fast evolving field with tremendous clinical potential.
Collapse
Affiliation(s)
- Z Wu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - N V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.
| |
Collapse
|
17
|
Yu S, Li A, Liu Q, Yuan X, Xu H, Jiao D, Pestell RG, Han X, Wu K. Recent advances of bispecific antibodies in solid tumors. J Hematol Oncol 2017; 10:155. [PMID: 28931402 PMCID: PMC5607507 DOI: 10.1186/s13045-017-0522-z] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 09/01/2017] [Indexed: 01/04/2023] Open
Abstract
Cancer immunotherapy is the most exciting advancement in cancer therapy. Similar to immune checkpoint blockade and chimeric antigen receptor T cell (CAR-T), bispecific antibody (BsAb) is attracting more and more attention as a novel strategy of antitumor immunotherapy. BsAb not only offers an effective linkage between therapeutics (e.g., immune effector cells, radionuclides) and targets (e.g., tumor cells) but also simultaneously blocks two different oncogenic mediators. In recent decades, a variety of BsAb formats have been generated. According to the structure of Fc domain, BsAb can be classified into two types: IgG-like format and Fc-free format. Among these formats, bispecific T cell engagers (BiTEs) and triomabs are commonly investigated. BsAb has achieved an exciting breakthrough in hematological malignancies and promising outcome in solid tumor as showed in various clinical trials. In this review, we focus on the preclinical experiments and clinical studies of epithelial cell adhesion molecule (EpCAM), human epidermal growth factor receptor (HER) family, carcinoembryonic antigen (CEA), and prostate-specific membrane antigen (PSMA) related BsAbs in solid tumors, as well as discuss the challenges and corresponding approaches in clinical application.
Collapse
Affiliation(s)
- Shengnan Yu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Anping Li
- Department of Interventional Radiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Qian Liu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Xun Yuan
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Hanxiao Xu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Dechao Jiao
- Department of Interventional Radiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Richard G Pestell
- Pennsylvania Center for Cancer and Regenerative Medicine, Wynnewood, PA, 19096, USA
| | - Xinwei Han
- Department of Interventional Radiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China.
| |
Collapse
|
18
|
Ranji P, Salmani Kesejini T, Saeedikhoo S, Alizadeh AM. Targeting cancer stem cell-specific markers and/or associated signaling pathways for overcoming cancer drug resistance. Tumour Biol 2016; 37:13059-13075. [DOI: 10.1007/s13277-016-5294-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/18/2016] [Indexed: 02/07/2023] Open
|
19
|
Subramanian N, Akilandeswari B, Bhutra A, Alameen M, Vetrivel U, Khetan V, Kanwar RK, Kanwar JR, Krishnakumar S. Targeting CD44, ABCG2 and CD133 markers using aptamers: in silico analysis of CD133 extracellular domain 2 and its aptamer. RSC Adv 2016. [DOI: 10.1039/c5ra27072c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Truncated CSC marker aptamers penetrate tumor spheres and inhibits cell proliferation; a bioinformatics approach to decipher their structural interactions.
Collapse
Affiliation(s)
- Nithya Subramanian
- Department of Nanobiotechnology
- Vision Research Foundation
- Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology
- Chennai – 600006
- India
| | - Balachandran Akilandeswari
- Department of Nanobiotechnology
- Vision Research Foundation
- Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology
- Chennai – 600006
- India
| | - Anjali Bhutra
- Department of Nanobiotechnology
- Vision Research Foundation
- Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology
- Chennai – 600006
- India
| | - Mohamed Alameen
- Centre for Bioinformatics
- Vision Research Foundation
- Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology
- Chennai – 600006
- India
| | - Umashankar Vetrivel
- Centre for Bioinformatics
- Vision Research Foundation
- Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology
- Chennai – 600006
- India
| | - Vikas Khetan
- Departments of Ocular Oncology and Vitreoretina
- Medical Research Foundation
- Sankara Nethralaya
- Chennai – 600006
- India
| | - Rupinder K. Kanwar
- Nanomedicine Laboratory of Immunology and Molecular Biomedical Research (NLIMBR)
- School of Medicine (SoM)
- Centre for Molecular and Medical Research (C-MMR) Strategic Research Centre
- Faculty of Health
- Deakin University
| | - Jagat R. Kanwar
- Nanomedicine Laboratory of Immunology and Molecular Biomedical Research (NLIMBR)
- School of Medicine (SoM)
- Centre for Molecular and Medical Research (C-MMR) Strategic Research Centre
- Faculty of Health
- Deakin University
| | - Subramanian Krishnakumar
- Department of Nanobiotechnology
- Vision Research Foundation
- Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology
- Chennai – 600006
- India
| |
Collapse
|
20
|
Blaudszun AR, Moldenhauer G, Schneider M, Philippi A. A photosensitizer delivered by bispecific antibody redirected T lymphocytes enhances cytotoxicity against EpCAM-expressing carcinoma cells upon light irradiation. J Control Release 2014; 197:58-68. [PMID: 25449805 DOI: 10.1016/j.jconrel.2014.10.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 09/05/2014] [Accepted: 10/28/2014] [Indexed: 01/19/2023]
Abstract
Recently conducted clinical trials have provided impressive evidence that chemotherapy resistant metastatic melanoma and several hematological malignancies can be cured using adoptive T cell therapy or T cell-recruiting bispecific antibodies. However, a significant fraction of patients did not benefit from these treatments. Here we have evaluated the feasibility of a novel combination therapy which aims to further enhance the killing potential of bispecific antibody-redirected T lymphocytes by using these cells as targeted delivery system for photosensitizing agents. For a first in vitro proof-of-concept study, ex vivo activated human donor T cells were loaded with a poly(styrene sulfonate) (PSS)-complex of the model photosensitizer 5,10,15,20-tetrakis(3-hydroxyphenyl)porphyrin (mTHPP). In the absence of light and when loading with the water-soluble PSS/mTHPP-complex occurred at a tolerable concentration, viability and cytotoxic function of loaded T lymphocytes were not impaired. When "drug-enhanced" T cells were co-cultivated with EpCAM-expressing human carcinoma cells, mTHPP was transferred to target cells. Notably, in the presence of a bispecific antibody, which cross-links effector and target cells thereby inducing the cytolytic activity of cytotoxic T lymphocytes, significantly more photosensitizer was transferred. Consequently, upon irradiation of co-cultures, redirected drug-loaded T cells were more effective in killing A549 lung and SKOV-3 ovarian carcinoma cells than retargeted unloaded T lymphocytes. Particularly, the additive approach using redirected unloaded T cells in combination with appropriate amounts of separately applied PSS/mTHPP was less efficient as well. Thus, by loading T lymphocytes with a stimulus-sensitive anti-cancer drug, we were able to enhance the cytotoxic capacity of carrier cells. Photosensitizer boosted T cells could open new perspectives for adoptive T cell therapy as well as targeted photodynamic therapy.
Collapse
Affiliation(s)
- André-René Blaudszun
- Environment and Bio Group, Korea Institute of Science and Technology (KIST) Europe Forschungsgesellschaft mbH, Saarland University, Campus E7 1, Saarbrücken D-66123, Germany.
| | - Gerhard Moldenhauer
- Department of Translational Immunology, German Cancer Research Center and National Center for Tumor Diseases, Heidelberg D-69120, Germany
| | - Marc Schneider
- Department of Pharmaceutics and Biopharmacy, Philipps-University, Ketzerbach 63, Marburg D-35037, Germany
| | - Anja Philippi
- Environment and Bio Group, Korea Institute of Science and Technology (KIST) Europe Forschungsgesellschaft mbH, Saarland University, Campus E7 1, Saarbrücken D-66123, Germany
| |
Collapse
|
21
|
Jhaveri DT, Zheng L, Jaffee EM. Specificity delivers: therapeutic role of tumor antigen-specific antibodies in pancreatic cancer. Semin Oncol 2014; 41:559-75. [PMID: 25440603 DOI: 10.1053/j.seminoncol.2014.07.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is among the most deadly cancers with less than 5% of the patients living beyond 5 years post-diagnosis. Lack of early diagnostic biomarkers and resistance to current therapies help explain these disappointing numbers. Thus, more effective and better-targeted therapies are needed quickly. Monoclonal antibodies offer an attractive alternative targeted therapy option for PDA because they are highly specific and potent. However, currently available monoclonal antibody therapies for PDA are still in their infancy with a low success rate and low likelihood of being approved. The challenges faced by these therapies include the following: lack of predictive and response biomarkers, unfavorable safety profiles, expression of targets not restricted to the cancer cells, flawed preclinical model systems, drug resistance, and PDA's complex nature. Additionally, discovery of novel PDA-specific antigen targets, present on the cell surface or in the extracellular matrix, is needed. Predictive and response markers also need to be determined for PDA patient subgroups so that the most appropriate effective therapy can be delivered. Serologic approaches, recombinant antibody-producing technologies, and advances in antibody engineering techniques will help to identify these predictive biomarkers and aid in the development of new therapeutic antibodies. A combinatorial approach simultaneously targeting antigens on the PDA cell, stroma, and immunosuppressive cells should be employed.
Collapse
Affiliation(s)
- Darshil T Jhaveri
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Oncology, the Sidney Kimmel Comprehensive Cancer Center and the Skip Viragh Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Lei Zheng
- Department of Oncology, the Sidney Kimmel Comprehensive Cancer Center and the Skip Viragh Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD.
| | - Elizabeth M Jaffee
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Oncology, the Sidney Kimmel Comprehensive Cancer Center and the Skip Viragh Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD.
| |
Collapse
|
22
|
Delgiorno KE, Hall JC, Takeuchi KK, Pan FC, Halbrook CJ, Washington MK, Olive KP, Spence JR, Sipos B, Wright CVE, Wells JM, Crawford HC. Identification and manipulation of biliary metaplasia in pancreatic tumors. Gastroenterology 2014; 146:233-44.e5. [PMID: 23999170 PMCID: PMC3870045 DOI: 10.1053/j.gastro.2013.08.053] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 08/02/2013] [Accepted: 08/22/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Metaplasias often have characteristics of developmentally related tissues. Pancreatic metaplastic ducts are usually associated with pancreatitis and pancreatic ductal adenocarcinoma. The tuft cell is a chemosensory cell that responds to signals in the extracellular environment via effector molecules. Commonly found in the biliary tract, tuft cells are absent from normal murine pancreas. Using the aberrant appearance of tuft cells as an indicator, we tested if pancreatic metaplasia represents transdifferentiation to a biliary phenotype and what effect this has on pancreatic tumorigenesis. METHODS We analyzed pancreatic tissue and tumors that developed in mice that express an activated form of Kras (Kras(LSL-G12D/+);Ptf1a(Cre/+) mice). Normal bile duct, pancreatic duct, and tumor-associated metaplasias from the mice were analyzed for tuft cell and biliary progenitor markers, including SOX17, a transcription factor that regulates biliary development. We also analyzed pancreatic tissues from mice expressing transgenic SOX17 alone (ROSA(tTa/+);Ptf1(CreERTM/+);tetO-SOX17) or along with activated Kras (ROSAtT(a/+);Ptf1a(CreERTM/+);tetO-SOX17;Kras(LSL-G12D;+)). RESULTS Tuft cells were frequently found in areas of pancreatic metaplasia, decreased throughout tumor progression, and absent from invasive tumors. Analysis of the pancreatobiliary ductal systems of mice revealed tuft cells in the biliary tract but not the normal pancreatic duct. Analysis for biliary markers revealed expression of SOX17 in pancreatic metaplasia and tumors. Pancreas-specific overexpression of SOX17 led to ductal metaplasia along with inflammation and collagen deposition. Mice that overexpressed SOX17 along with Kras(G12D) had a greater degree of transformed tissue compared with mice expressing only Kras(G12D). Immunofluorescence analysis of human pancreatic tissue arrays revealed the presence of tuft cells in metaplasia and early-stage tumors, along with SOX17 expression, consistent with a biliary phenotype. CONCLUSIONS Expression of Kras(G12D) and SOX17 in mice induces development of metaplasias with a biliary phenotype containing tuft cells. Tuft cells express a number of tumorigenic factors that can alter the microenvironment. Expression of SOX17 induces pancreatitis and promotes Kras(G12D)-induced tumorigenesis in mice.
Collapse
Affiliation(s)
- Kathleen E Delgiorno
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York; Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
| | - Jason C Hall
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida; Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York
| | | | - Fong Cheng Pan
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Christopher J Halbrook
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida; Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York
| | - M Kay Washington
- Department of Pathology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kenneth P Olive
- Departments of Medicine and Pathology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| | - Jason R Spence
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Bence Sipos
- Department of Pathology, University Hospital Tubingen, Tubingen, Germany
| | - Christopher V E Wright
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - James M Wells
- Department of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Howard C Crawford
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York; Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida.
| |
Collapse
|
23
|
Abstract
Despite significant advances in surgery, radiotherapy and chemotherapy to treat prostate cancer (CaP), many patients die of secondary disease (metastases). Current therapeutic approaches are limited, and there is no cure for metastatic castration-resistant prostate cancer (CRPC). Epithelial cell adhesion molecule (EpCAM, also known as CD326) is a transmembrane glycoprotein that is highly expressed in rapidly proliferating carcinomas and plays an important role in the prevention of cell-cell adhesion, cell signalling, migration, proliferation and differentiation. Stably and highly expressed EpCAM has been found in primary CaP tissues, effusions and CaP metastases, making it an ideal candidate of tumour-associated antigen to detect metastasis of CaP cells in the circulation as well as a promising therapeutic target to control metastatic CRPC disease. In this review, we discuss the implications of the newly identified roles of EpCAM in terms of its diagnostic and metastatic relevance to CaP. We also summarize EpCAM expression in human CaP and EpCAM-mediated signalling pathways in cancer metastasis. Finally, emerging and innovative approaches to the management of the disease and expanding potential therapeutic applications of EpCAM for targeted strategies in future CaP therapy will be explored.
Collapse
|
24
|
Ning X, Shu J, Du Y, Ben Q, Li Z. Therapeutic strategies targeting cancer stem cells. Cancer Biol Ther 2013; 14:295-303. [PMID: 23358473 DOI: 10.4161/cbt.23622] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Increasing studies have demonstrated a small proportion of cancer stem cells (CSCs) exist in the cancer cell population. CSCs have powerful self-renewal capacity and tumor-initiating ability and are resistant to chemotherapy and radiation. Conventional anticancer therapies kill the rapidly proliferating bulk cancer cells but spare the relatively quiescent CSCs, which cause cancer recurrence. So it is necessary to develop therapeutic strategies acting specifically on CSCs. In recent years, studies have shown that therapeutic agents such as metformin, salinomycin, DECA-14, rapamycin, oncostatin M (OSM), some natural compounds, oncolytic viruses, microRNAs, cell signaling pathway inhibitors, TNF-related apoptosis inducing ligand (TRAIL), interferon (IFN), telomerase inhibitors, all-trans retinoic acid (ATRA) and monoclonal antibodies can suppress the self-renewal of CSCs in vitro and in vivo. A combination of these agents and conventional chemotherapy drugs can significantly inhibit tumor growth, metastasis and recurrence. These strategies targeting CSCs may bring new hopes to cancer therapy.
Collapse
Affiliation(s)
- Xiaoyan Ning
- Department of Gastroenterology, Changhai Hospital of Second Military Medical University, Shanghai, China
| | | | | | | | | |
Collapse
|
25
|
Connor JP, Cristea MC, Lewis NL, Lewis LD, Komarnitsky PB, Mattiacci MR, Felder M, Stewart S, Harter J, Henslee-Downey J, Kramer D, Neugebauer R, Stupp R. A phase 1b study of humanized KS-interleukin-2 (huKS-IL2) immunocytokine with cyclophosphamide in patients with EpCAM-positive advanced solid tumors. BMC Cancer 2013; 13:20. [PMID: 23320927 PMCID: PMC3600662 DOI: 10.1186/1471-2407-13-20] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 01/11/2013] [Indexed: 11/10/2022] Open
Abstract
Background Humanized KS-interleukin-2 (huKS-IL2), an immunocytokine with specificity for epithelial cell adhesion molecule (EpCAM), has demonstrated favorable tolerability and immunologic activity as a single agent. Methods Phase 1b study in patients with EpCAM-positive advanced solid tumors to determine the maximum tolerated dose (MTD) and safety profile of huKS-IL2 in combination with low-dose cyclophosphamide. Treatment consisted of cyclophosphamide (300 mg/m2 on day 1), and escalating doses of huKS-IL2 (0.5–4.0 mg/m2 IV continuous infusion over 4 hours) on days 2, 3, and 4 of each 21-day cycle. Safety, pharmacokinetic profile, immunogenicity, anti-tumor and biologic activity were evaluated. Results Twenty-seven patients were treated for up to 6 cycles; 26 were evaluable for response. The MTD of huKS-IL2 in combination with 300 mg/m2 cyclophosphamide was 3.0 mg/m2. At higher doses, myelosuppression was dose-limiting. Transient lymphopenia was the most common grade 3/4 adverse event (AE). Other significant AEs included hypotension, hypophosphatemia, and increase in serum creatinine. All patients recovered from these AEs. The huKS-IL2 exposure was dose-dependent, but not dose-proportional, accumulation was negligible, and elimination half-life and systemic clearance were independent of dose and time. Most patients had a transient immune response to huKS-IL2. Immunologic activity was observed at all doses. Ten patients (38%) had stable disease as best response, lasting for ≥ 4 cycles in 3 patients. Conclusion The combination of huKS-IL2 with low-dose cyclophosphamide was well tolerated. Although no objective responses were observed, the combination showed evidence of immunologic activity and 3 patients showed stable disease for ≥ 4 cycles. Trial registration http://NCT00132522
Collapse
|
26
|
Abstract
Understanding the genetic and molecular mechanisms of ovarian cancer has been the focus of research efforts working toward the greater goal of improving cancer therapy for patients with residual disease after initial treatment with conventional surgery and neoadjuvant chemotherapy. The focus of this review will be centered on new therapeutic strategies based on Cancer Stem Cells studies of chemoresistant subpopulations, the prevention of metastasis, and individualized therapy in order to find the most successful combination of treatments to effectively treat human ovarian cancer. We reviewed recent literature (1993-2011) of novel treatment approaches to ovarian cancer stem cells. As the focus of ovarian cancer investigation has centered on the cancer stem cell model and the complexities that it presents in the development of effective treatments, the future of treating ovarian cancer lies in utilizing individualized treatment systems that include enhancing existing treatments, aiming for novel therapy targets, managing the plasticity of stem cells to induce cellular differentiation, and regulating oncogenic signaling pathways.
Collapse
|
27
|
Alama A, Orengo AM, Ferrini S, Gangemi R. Targeting cancer-initiating cell drug-resistance: a roadmap to a new-generation of cancer therapies? Drug Discov Today 2012; 17:435-42. [DOI: 10.1016/j.drudis.2011.02.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Accepted: 02/04/2011] [Indexed: 01/20/2023]
|
28
|
Schmetzer O, Moldenhauer G, Nicolaou A, Schlag P, Riesenberg R, Pezzutto A. Detection of circulating tumor-associated antigen depends on the domains recognized by the monoclonal antibodies used: N-terminal trimmed EpCAM-levels are much higher than untrimmed forms. Immunol Lett 2012; 143:184-92. [DOI: 10.1016/j.imlet.2012.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 01/29/2012] [Accepted: 02/14/2012] [Indexed: 10/28/2022]
|
29
|
Moldenhauer G, Salnikov AV, Lüttgau S, Herr I, Anderl J, Faulstich H. Therapeutic potential of amanitin-conjugated anti-epithelial cell adhesion molecule monoclonal antibody against pancreatic carcinoma. J Natl Cancer Inst 2012; 104:622-34. [PMID: 22457476 DOI: 10.1093/jnci/djs140] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Human epithelial cell adhesion molecule (EpCAM) is overexpressed in many cancers. Anti-EpCAM antibodies have shown promise in preclinical studies, but showed no tumor regression in a recent phase II clinical trial. Therefore, we generated a novel anti-EpCAM antibody-drug conjugate and assessed whether it showed enhanced antitumor effects. METHODS Chemical cross-linking was conducted to covalently conjugate α-amanitin, a toxin known to inhibit DNA transcription, with chiHEA125, a chimerized anti-EpCAM monoclonal antibody, to generate the antibody-drug conjugate α-amanitin-glutarate-chiHEA125 (chiHEA125-Ama). Antiproliferative activity of chiHEA125-Ama was tested in human pancreatic (BxPc-3 and Capan-1), colorectal (Colo205), breast (MCF-7), and bile duct (OZ) cancer cell lines in vitro using [(3)H]-thymidine incorporation assay. Antitumor activity of chiHEA125-Ama was assessed in vivo in immunocompromised mice bearing subcutaneous human BxPc-3 pancreatic carcinoma xenograft tumors (n = 66 mice). Cell proliferation and apoptosis were evaluated in xenograft tumors by immunohistochemistry. All statistical tests were two-sided. RESULTS In all cell lines, chiHEA125-Ama reduced cell proliferation (mean half maximal inhibitory concentration [IC(50)] = 2.5 × 10(-10) to 5.4 × 10(-12) M). A single dose of chiHEA125-Ama inhibited BxPc-3 xenograft tumor growth (chiHEA125 [control, n = 4 mice] vs. chiHEA125-Ama [n = 6 mice], dose of 15 mg/kg with respect to IgG and 50 μg/kg with respect to α-amanitin, mean relative increase in tumor volume on day 16 = 884% vs. -79%, difference = 963%, 95% CI = 582% to 1344%, P = .019). Two higher doses of chiHEA125-Ama (100 μg/kg with respect to α-amanitin), administered 1 week apart (n = 10 mice per group), led to complete tumor regression in nine of 10 (90%) mice compared with chiHEA125, during the observation period of 16 days; increased apoptosis and reduced cell proliferation were observed in mice treated with chiHEA125-Ama. CONCLUSION This preclinical study suggests that anti-EpCAM antibody conjugates with α-amanitin have the potential to be highly effective therapeutic agents for pancreatic carcinomas and various EpCAM-expressing malignancies.
Collapse
Affiliation(s)
- Gerhard Moldenhauer
- Department of Translational Immunology (D015), German Cancer Research Center, Im Neuenheimer Feld 280, Heidelberg 69120, Germany.
| | | | | | | | | | | |
Collapse
|
30
|
Mitra M, Kandalam M, Harilal A, Verma RS, Krishnan UM, Swaminathan S, Krishnakumar S. EpCAM is a putative stem marker in retinoblastoma and an effective target for T-cell-mediated immunotherapy. Mol Vis 2012; 18:290-308. [PMID: 22328825 PMCID: PMC3275636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Accepted: 01/26/2012] [Indexed: 11/04/2022] Open
Abstract
PURPOSE The molecular markers cluster of differentiation (CD)24, CD44, adenosine tri-phosphate (ATP) binding cassette protein G2 (ABCG2), and epithelial cell adhesion molecule (EpCAM) are widely used, individually or in combination, to characterize some types of cancer stem cells. In this study we characterized the EpCAM+ retinoblastoma (RB) cells for their cancer stem-like properties in vitro. Additionally, we targeted RB tumor cells via redirecting T cells using bispecific EpCAM×CD3 antibody. METHODS Flow cytometry was used to study the co-expression of EpCAM with putative cancer stem cell markers, such as CD44, CD24, and ABCG2, in RB primary tumors. In vitro methyl thiazol tetrazolium (MTT) assay, invasion assay, and neurosphere formation assay were performed to characterize EpCAM+ cells for their cancer stem/progenitor cell-like properties. We assessed the in vitro efficacy of bispecific EpCAM×CD3 antibody on RB tumor cell proliferation and validated the results by evaluating effector cytokine production in the culture medium with the ELISA method. RESULTS EpCAM was co-expressed with all cancer stem cell markers (CD44, CD24, and ABCG2) in primary RB tumors. EpCAM+ cells showed significantly higher proliferative invasive potential and neurosphere formation in vitro compared to EpCAM⁻ Y79 cells. EpCAM+ cells showed higher β-catenin expression compared to EpCAM- cells. EpCAM×CD3 significantly retarded proliferation of RB primary tumor cells. EpCAM×CD3 effectively induced the secretion of effector cytokines, such as interferon (IFN)-γ, tumor necrosis factor (TNF)-α, interleukin (IL)-10, IL-2, and transforming growth factor (TGF)-β1, and also perforin levels by pre-activated lymphocytes. CONCLUSIONS EpCAM might be a novel cancer stem cell marker in RB. EpCAM×CD3 antibody redirecting T cells to attack RB tumor cells may prove effective in RB management. Further preclinical studies are needed to confirm the initial findings of our study.
Collapse
Affiliation(s)
- Moutushy Mitra
- L&T Department of Ocular Pathology, Vision Research Foundation, Tamil Nadu, India,CeNTAB, SASTRA University, Tanjore, India
| | | | - Anju Harilal
- L&T Department of Ocular Pathology, Vision Research Foundation, Tamil Nadu, India
| | - Rama Shenkar Verma
- Department of Biotechnology, Indian Institute of Technology (IIT), Madras, India
| | | | | | | |
Collapse
|
31
|
Groth A, Salnikov AV, Ottinger S, Gladkich J, Liu L, Kallifatidis G, Salnikova O, Ryschich E, Giese N, Giese T, Momburg F, Büchler MW, Moldenhauer G, Herr I. New Gene-Immunotherapy Combining TRAIL-Lymphocytes and EpCAMxCD3 Bispecific Antibody for Tumor Targeting. Clin Cancer Res 2012; 18:1028-38. [DOI: 10.1158/1078-0432.ccr-11-2767] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
32
|
Chen X, Lingala S, Khoobyari S, Nolta J, Zern MA, Wu J. Epithelial mesenchymal transition and hedgehog signaling activation are associated with chemoresistance and invasion of hepatoma subpopulations. J Hepatol 2011; 55:838-45. [PMID: 21334406 PMCID: PMC3177032 DOI: 10.1016/j.jhep.2010.12.043] [Citation(s) in RCA: 169] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Revised: 12/21/2010] [Accepted: 12/22/2010] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Our previous studies showed that CD133, EpCAM, and aldehyde dehydrogenase (ALDH) are useful markers to identify cancer stem cells (CSCs) in hepatocellular carcinoma (HCC) tissues. The present study aims to evaluate chemosensitivity and invasion capability of HCC based on CSC marker profiles, and to explore the underlying molecular mechanisms. METHODS Hepatoma cell lines were separated into subpopulations according to CD133, EpCAM, and ALDH expression profiles. Epithelial mesenchymal transition (EMT) and hedgehog (Hh) signaling were examined to identify their links with chemoresistance and aggressive invasion. RESULTS Well-differentiated cell lines were positive for CD133(+)/ALDH(high) and CD133(+)/EpCAM(+) at 1.5-15% and 2.3-8.3%; whereas, poorly-differentiated cells were almost all negative for these markers. FACS-enriched CD133(+)/ALDH(high) and CD133(+)/EpCAM(+) Hep3B and Huh-7 cells formed more spheroids in vitro. CD133(-)/ALDH(low) HLE cells were more resistant to cisplatin, doxorubicin or sorafenib than their positive counterparts. CD133(-)/EpCAM(-) Huh-7 cells or CD133(-)/ALDH(-) HLE cells exhibited a higher invasion rate than their positive counterparts. HLE and HLF cells acquired EMT in double negative subpopulations. Hh activity in Huh-7 CD133(-)/EpCAM(-) cells was higher than in their positive counterparts, and the inhibition of Hh activity by cyclopamine resulted in reduced cell proliferation. CONCLUSIONS Well-differentiated CD133(+)/ALDH(high) or CD133(+)/EpCAM(+) cells appear to be a CSC/initiating subpopulation; whereas, in poorly-differentiated hepatoma cells, EMT and enhanced hedgehog signaling activity may be responsible for their chemoresistance and invasion. These findings underscore the significance of EMT and enhanced Hh signaling in liver cancer stem or initiating cells.
Collapse
Affiliation(s)
- Xiaoli Chen
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of California Davis Medical Center, Sacramento, CA 95817
| | - Shilpa Lingala
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of California Davis Medical Center, Sacramento, CA 95817
| | - Shiva Khoobyari
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of California Davis Medical Center, Sacramento, CA 95817
| | - Jan Nolta
- Stem Cell Program, University of California, Davis Medical Center, Sacramento, CA 95817
| | - Mark A. Zern
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of California Davis Medical Center, Sacramento, CA 95817
| | - Jian Wu
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of California Davis Medical Center, Sacramento, CA 95817
- Stem Cell Program, University of California, Davis Medical Center, Sacramento, CA 95817
- Cancer Center, University of California, Davis Medical Center, Sacramento, CA 95817
| |
Collapse
|
33
|
Patel SA, Dave MA, Murthy RG, Helmy KY, Rameshwar P. Metastatic breast cancer cells in the bone marrow microenvironment: novel insights into oncoprotection. Oncol Rev 2011; 5:93-102. [PMID: 21776337 PMCID: PMC3138628 DOI: 10.1007/s12156-010-0071-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Among all cancers, malignancies of the breast are the second leading cause of cancer death in the United States after carcinoma of the lung. One of the major factors considered when assessing the prognosis of breast cancer patients is whether the tumor has metastasized to distant organs. Although the exact phenotype of the malignant cells responsible for metastasis and dormancy is still unknown, growing evidence has revealed that they may have stem cell-like properties that may account for resistance to chemotherapy and radiation. One process that has been attributed to primary tumor metastasis is the epithelial-to-mesenchymal transition. In this review, we specifically discuss breast cancer dissemination to the bone marrow and factors that ultimately serve to shelter and promote tumor growth, including the complex relationship between mesenchymal stem cells (MSCs) and various aspects of the immune system, carcinoma-associated fibroblasts, and the diverse components of the tumor microenvironment. A better understanding of the journey from the primary tumor site to the bone marrow and subsequently the oncoprotective role of MSCs and other factors within that microenvironment can potentially lead to development of novel therapeutic targets.
Collapse
Affiliation(s)
- Shyam A. Patel
- Division of Hematology/Oncology, Department of Medicine, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, MSB, Room E-579, 185 South Orange Avenue, Newark, NJ 07103, USA. Graduate School of Biomedical Sciences, University of Medicine and Dentistry of New Jersey, Newark, NJ, USA
| | - Meneka A. Dave
- Division of Hematology/Oncology, Department of Medicine, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, MSB, Room E-579, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Raghav G. Murthy
- Division of Hematology/Oncology, Department of Medicine, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, MSB, Room E-579, 185 South Orange Avenue, Newark, NJ 07103, USA
| | - Karim Y. Helmy
- Division of Hematology/Oncology, Department of Medicine, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, MSB, Room E-579, 185 South Orange Avenue, Newark, NJ 07103, USA. Graduate School of Biomedical Sciences, University of Medicine and Dentistry of New Jersey, Newark, NJ, USA
| | - Pranela Rameshwar
- Division of Hematology/Oncology, Department of Medicine, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, MSB, Room E-579, 185 South Orange Avenue, Newark, NJ 07103, USA
| |
Collapse
|
34
|
Chames P, Kerfelec B, Baty D. Therapeutic antibodies for the treatment of pancreatic cancer. ScientificWorldJournal 2010; 10:1107-20. [PMID: 20563534 PMCID: PMC2925140 DOI: 10.1100/tsw.2010.103] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Pancreatic cancer is a devastating disease with the worst mortality rate and an overall 5-year survival rate lower than 5%. In the U.S., this disease is the fourth leading cause of death and represents 6% of all cancer-related deaths. Gemcitabine, the current standard first-line treatment, offers marginal benefits to patients in terms of symptom control and prolongation of life. Since 1996, about 20 randomized phase III trials have been performed to improve the efficacy of gemcitabine, with little success regarding a significant improvement in survival outcomes. The need for novel therapeutic strategies, such as target therapy, is obvious. Monoclonal antibodies have finally come of age as therapeutics and several molecules are now approved for cancer therapies. This review aims to give a general view on the clinical results obtained so far by antibodies for the treatment of pancreatic cancer and describes the most promising avenues toward a significant improvement in the treatment of this frustrating disease.
Collapse
|
35
|
Yu X, Zhang Y, Chen C, Yao Q, Li M. Targeted drug delivery in pancreatic cancer. Biochim Biophys Acta Rev Cancer 2009; 1805:97-104. [PMID: 19853645 DOI: 10.1016/j.bbcan.2009.10.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Revised: 10/07/2009] [Accepted: 10/11/2009] [Indexed: 12/16/2022]
Abstract
Effective drug delivery in pancreatic cancer treatment remains a major challenge. Because of the high resistance to chemo and radiation therapy, the overall survival rate for pancreatic cancer is extremely low. Recent advances in drug delivery systems hold great promise for improving cancer therapy. Using liposomes, nanoparticles, and carbon nanotubes to deliver cancer drugs and other therapeutic agents such as siRNA, suicide gene, oncolytic virus, small molecule inhibitor, and antibody has been a success in recent preclinical trials. However, how to improve the specificity and stability of the delivered drug using ligand or antibody directed delivery represent a major problem. Therefore, developing novel, specific, tumor-targeted drug delivery systems is urgently needed for this terrible disease. This review summarizes the current progress on targeted drug delivery in pancreatic cancer and provides important information on potential therapeutic targets for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Xianjun Yu
- Michael E. DeBakey Department of Surgery, Molecular Surgeon Research Center, Elkins Pancreas Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|