1
|
Zhao Y, Ni Q, Zhang W, Yu S. Progress in reeducating tumor-associated macrophages in tumor microenvironment. Discov Oncol 2024; 15:312. [PMID: 39060648 PMCID: PMC11282027 DOI: 10.1007/s12672-024-01186-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024] Open
Abstract
Malignant tumor, one of the most threatening diseases to human health, has been comprehensively treated with surgery, radiotherapy, chemotherapy and targeted therapy, but the prognosis has not always been ideal. In the past decade, immunotherapy has shown increased efficacy in tumor treatment; however, for immunotherapy to achieve its fullest potential, obstacles are to be conquered, among which tumor microenvironment (TME) has been widely investigated. In remodeling the tumor immune microenvironment to inhibit tumor progression, macrophages, as the most abundant innate immune population, play an irreplaceable role in the immune response. Therefore, how to remodel TME and alter the recruitment and polarization status of tumor-associated macrophages (TAM) has been of wide interest. In this context, nanoparticles, photodynamic therapy and other therapeutic approaches capable of affecting macrophage polarization have emerged. In this paper, we categorize and organize the existing means and methods for reprogramming TAM to provide ideas for clinical application of novel tumor-related therapies.
Collapse
Affiliation(s)
- Yiming Zhao
- Department of Gastrointestinal Surgery, The Third Hospital of Hebei Medical University, No.139, Ziqiang Road, Qiaoxi District, Shijiazhuang, 050051, China
| | - Qianyang Ni
- Department of Gastrointestinal Surgery, The Third Hospital of Hebei Medical University, No.139, Ziqiang Road, Qiaoxi District, Shijiazhuang, 050051, China
| | - Weijian Zhang
- Department of Gastrointestinal Surgery, The Third Hospital of Hebei Medical University, No.139, Ziqiang Road, Qiaoxi District, Shijiazhuang, 050051, China
| | - Suyang Yu
- Department of Gastrointestinal Surgery, The Third Hospital of Hebei Medical University, No.139, Ziqiang Road, Qiaoxi District, Shijiazhuang, 050051, China.
| |
Collapse
|
2
|
Ren Y, Wang M, Yuan H, Wang Z, Yu L. A novel insight into cancer therapy: Lipid metabolism in tumor-associated macrophages. Int Immunopharmacol 2024; 135:112319. [PMID: 38801810 DOI: 10.1016/j.intimp.2024.112319] [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: 03/19/2024] [Revised: 05/07/2024] [Accepted: 05/19/2024] [Indexed: 05/29/2024]
Abstract
The tumor immune microenvironment (TIME) can limit the effectiveness and often leads to significant side effects of conventional cancer therapies. Consequently, there is a growing interest in identifying novel targets to enhance the efficacy of targeted cancer therapy. More research indicates that tumor-associated macrophages (TAMs), originating from peripheral blood monocytes generated from bone marrow myeloid progenitor cells, play a crucial role in the tumor microenvironment (TME) and are closely associated with resistance to traditional cancer therapies. Lipid metabolism alterations have been widely recognized as having a significant impact on tumors and their immune microenvironment. Lipids, lipid derivatives, and key substances in their metabolic pathways can influence the carcinogenesis and progression of cancer cells by modulating the phenotype, function, and activity of TAMs. Therefore, this review focuses on the reprogramming of lipid metabolism in cancer cells and their immune microenvironment, in which the TAMs are especially concentrated. Such changes impact TAMs activation and polarization, thereby affecting the tumor cell response to treatment. Furthermore, the article explores the potential of targeting the lipid metabolism of TAMs as a supplementary approach to conventional cancer therapies. It reviews and evaluates current strategies for enhancing efficacy through TAMs' lipid metabolism and proposes new lipid metabolism targets as potential synergistic options for chemo-radiotherapy and immunotherapy. These efforts aim to stimulate further research in this area.
Collapse
Affiliation(s)
- Yvxiao Ren
- Department of Radiotherapy, Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Mingjie Wang
- Department of Radiotherapy, Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, People's Republic of China
| | - Hanghang Yuan
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, People's Republic of China
| | - Zhicheng Wang
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, People's Republic of China
| | - Lei Yu
- Department of Radiotherapy, Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China.
| |
Collapse
|
3
|
Liang C, Zhang Y, Wang S, Jiao W, Guo J, Zhang N, Liu X. Nanomaterials in modulating tumor-associated macrophages and enhancing immunotherapy. J Mater Chem B 2024; 12:4809-4823. [PMID: 38695349 DOI: 10.1039/d4tb00230j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
Tumor-associated macrophages (TAMs) are predominantly present in the tumor microenvironment (TME) and play a crucial role in shaping the efficacy of tumor immunotherapy. These TAMs primarily exhibit a tumor-promoting M2-like phenotype, which is associated with the suppression of immune responses and facilitation of tumor progression. Interestingly, recent research has highlighted the potential of repolarizing TAMs from an M2 to a pro-inflammatory M1 status-a shift that has shown promise in impeding tumor growth and enhancing immune responsiveness. This concept is particularly intriguing as it offers a new dimension to cancer therapy by targeting the tumor microenvironment, which is a significant departure from traditional approaches that focus solely on tumor cells. However, the clinical application of TAM-modulating agents is often challenged by issues such as insufficient tumor accumulation and off-target effects, limiting their effectiveness and safety. In this regard, nanomaterials have emerged as a novel solution. They serve a dual role: as delivery vehicles that can enhance the accumulation of therapeutic agents in the tumor site and as TAM-modulators. This dual functionality of nanomaterials is a significant advancement as it addresses the key limitations of current TAM-modulating strategies and opens up new avenues for more efficient and targeted therapies. This review provides a comprehensive overview of the latest mechanisms and strategies involving nanomaterials in modulating macrophage polarization within the TME. It delves into the intricate interactions between nanomaterials and macrophages, elucidating how these interactions can be exploited to drive macrophage polarization towards a phenotype that is more conducive to anti-tumor immunity. Additionally, the review explores the burgeoning field of TAM-associated nanomedicines in combination with tumor immunotherapy. This combination approach is particularly promising as it leverages the strengths of both nanomedicine and immunotherapy, potentially leading to synergistic effects in combating cancer.
Collapse
Affiliation(s)
- Chen Liang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, The College of Life Sciences & School of Medicine, Northwest University, Xi'an, Shaanxi 710069, China.
| | - Yihan Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, Shaanxi 710127, China
| | - Siyao Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, The College of Life Sciences & School of Medicine, Northwest University, Xi'an, Shaanxi 710069, China.
| | - Wangbo Jiao
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, Shaanxi 710127, China
| | - Jingyi Guo
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, The College of Life Sciences & School of Medicine, Northwest University, Xi'an, Shaanxi 710069, China.
| | - Nan Zhang
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Xiaoli Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, The College of Life Sciences & School of Medicine, Northwest University, Xi'an, Shaanxi 710069, China.
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
4
|
Dowaidar M. Guidelines for the role of autophagy in drug delivery vectors uptake pathways. Heliyon 2024; 10:e30238. [PMID: 38707383 PMCID: PMC11066435 DOI: 10.1016/j.heliyon.2024.e30238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/22/2024] [Accepted: 04/22/2024] [Indexed: 05/07/2024] Open
Abstract
The process of autophagy refers to the intracellular absorption of cytoplasm (such as proteins, nucleic acids, tiny molecules, complete organelles, and so on) into the lysosome, followed by the breakdown of that cytoplasm. The majority of cellular proteins are degraded by a process called autophagy, which is both a naturally occurring activity and one that may be induced by cellular stress. Autophagy is a system that can save cells' integrity in stressful situations by restoring metabolic basics and getting rid of subcellular junk. This happens as a component of an endurance response. This mechanism may have an effect on disease, in addition to its contribution to the homeostasis of individual cells and tissues as well as the control of development in higher species. The main aim of this study is to discuss the guidelines for the role of autophagy in drug delivery vector uptake pathways. In this paper, we discuss the meaning and concept of autophagy, the mechanism of autophagy, the role of autophagy in drug delivery vectors, autophagy-modulating drugs, nanostructures for delivery systems of autophagy modulators, etc. Later in this paper, we talk about how to deliver chemotherapeutics, siRNA, and autophagy inducers and inhibitors. We also talk about how hard it is to make a drug delivery system that takes nanocarriers' roles as autophagy modulators into account.
Collapse
Affiliation(s)
- Moataz Dowaidar
- Bioengineering Department, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, 31261, Saudi Arabia
- Interdisciplinary Research Center for Hydrogen Technologies and Carbon Management, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, 31261, Saudi Arabia
- Biosystems and Machines Research Center, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, 31261, Saudi Arabia
| |
Collapse
|
5
|
Sesarman A, Luput L, Rauca VF, Patras L, Licarete E, Meszaros MS, Dume BR, Negrea G, Toma VA, Muntean D, Porfire A, Banciu M. Targeting of M2 macrophages with IL-13-functionalized liposomal prednisolone inhibits melanoma angiogenesis in vivo. J Liposome Res 2024:1-12. [PMID: 38379249 DOI: 10.1080/08982104.2024.2315452] [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: 07/31/2023] [Accepted: 01/31/2024] [Indexed: 02/22/2024]
Abstract
The intricate cooperation between cancer cells and nontumor stromal cells within melanoma microenvironment (MME) enables tumor progression and metastasis. We previously demonstrated that the interplay between tumor-associated macrophages (TAMs) and melanoma cells can be disrupted by using long-circulating liposomes (LCLs) encapsulating prednisolone phosphate (PLP) (LCL-PLP) that inhibited tumor angiogenesis coordinated by TAMs. In this study, our goal was to improve LCL specificity for protumor macrophages (M2-like (i.e., TAMs) macrophages) and to induce a more precise accumulation at tumor site by loading PLP into IL-13-conjugated liposomes (IL-13-LCL-PLP), since IL-13 receptor is overexpressed in this type of macrophages. The IL-13-LCL-PLP liposomal formulation was obtained by covalent attachment of thiolated IL-13 to maleimide-functionalized LCL-PLP. C57BL/6 mice bearing B16.F10 s.c melanoma tumors were used to investigate the antitumor action of LCL-PLP and IL-13-LCL-PLP. Our results showed that IL-13-LCL-PLP formulation remained stable in biological fluids after 24h and it was preferentially taken up by M2 polarized macrophages. IL-13-LCL-PLP induced strong tumor growth inhibition compared to nonfunctionalized LCL-PLP at the same dose, by altering TAMs-mediated angiogenesis and oxidative stress, limiting resistance to apoptosis and invasive features in MME. These findings suggest IL-13-LCL-PLP might become a promising delivery platform for chemotherapeutic agents in melanoma.
Collapse
Affiliation(s)
- Alina Sesarman
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Lavinia Luput
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Valentin-Florian Rauca
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
- Department of Dermatology and Allergology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Laura Patras
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Emilia Licarete
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
- Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences of Babes-Bolyai University, Cluj-Napoca, Romania
| | - Marta-Szilvia Meszaros
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Bogdan Razvan Dume
- Doctoral School in Integrative Biology, Faculty of Biology and Geology, "Babes-Bolyai" University, Cluj-Napoca, Romania
| | - Giorgiana Negrea
- Doctoral School in Integrative Biology, Faculty of Biology and Geology, "Babes-Bolyai" University, Cluj-Napoca, Romania
| | - Vlad-Alexandru Toma
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
- Department of Experimental Biology and Biochemistry, nstitute of Biological Research, branch of NIRDBS Bucharest, Cluj-Napoca, Romania
| | - Dana Muntean
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Hatieganu", Cluj-Napoca, Romania
| | - Alina Porfire
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Hatieganu", Cluj-Napoca, Romania
| | - Manuela Banciu
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| |
Collapse
|
6
|
Mehta P, Shende P. Dual role of autophagy for advancements from conventional to new delivery systems in cancer. Biochim Biophys Acta Gen Subj 2023; 1867:130430. [PMID: 37506854 DOI: 10.1016/j.bbagen.2023.130430] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023]
Abstract
Autophagy, a programmed cell-lysis mechanism, holds significant promise in the prevention and treatment of a wide range of conditions, including cancer, Alzheimer's, and Parkinson's disease. The successful utilization of autophagy modulation for therapeutic purposes hinges upon accurately determining the role of autophagy in disease progression, whether it acts as a cytotoxic or cytoprotective factor. This critical knowledge empowers scientists to effectively manipulate tumor sensitivity to anti-cancer therapies through autophagy modulation, while also circumventing drug resistance. However, conventional therapies face limitations such as low bioavailability, poor solubility, and a lack of controlled release mechanisms, hindering their clinical applicability. In this regard, innovative nanoplatforms including organic and inorganic systems have emerged as promising solutions to offer stimuli-responsive, theranostic-controlled drug delivery systems with active targeting and improved solubility. The review article explores a variety of organic nanoplatforms, such as lipid-based, polymer-based, and DNA-based systems, which incorporate autophagy-inhibiting drugs like hydroxychloroquine. By inhibiting the glycolytic pathway and depriving cells of essential nutrients, these platforms exhibit tumor-suppressive effects in advanced forms of cancer such as leukemia, colon cancer, and glioblastoma. Furthermore, metal-based, metal-oxide-based, silica-based, and quantum dot-based nanoplatforms selectively induce autophagy in tumors, leading to extensive cancer cell destruction. Additionally, this article discusses the current clinical status of autophagy-modulating drugs for cancer therapy with valuable insights of progress and potential of such approaches.
Collapse
Affiliation(s)
- Parth Mehta
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai, India
| | - Pravin Shende
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai, India.
| |
Collapse
|
7
|
Liu W, Wang B, Zhou M, Liu D, Chen F, Zhao X, Lu Y. Redox Dysregulation in the Tumor Microenvironment Contributes to Cancer Metastasis. Antioxid Redox Signal 2023; 39:472-490. [PMID: 37002890 DOI: 10.1089/ars.2023.0272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Significance: Redox dysregulation under pathological conditions results in excessive reactive oxygen species (ROS) accumulation, leading to oxidative stress and cellular oxidative damage. ROS function as a double-edged sword to modulate various types of cancer development and survival. Recent Advances: Emerging evidence has underlined that ROS impact the behavior of both cancer cells and tumor-associated stromal cells in the tumor microenvironment (TME), and these cells have developed complex systems to adapt to high ROS environments during cancer progression. Critical Issues: In this review, we integrated current progress regarding the impact of ROS on cancer cells and tumor-associated stromal cells in the TME and summarized how ROS production influences cancer cell behaviors. Then, we summarized the distinct effects of ROS during different stages of tumor metastasis. Finally, we discussed potential therapeutic strategies for modulating ROS for the treatment of cancer metastasis. Future Directions: Targeting the ROS regulation during cancer metastasis will provide important insights into the design of effective single or combinatorial cancer therapeutic strategies. Well-designed preclinical studies and clinical trials are urgently needed to understand the complex regulatory systems of ROS in the TME. Antioxid. Redox Signal. 39, 472-490.
Collapse
Affiliation(s)
- Wanning Liu
- College of Life Sciences, Northwest University, Xi'an, China
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Boda Wang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Mingzhen Zhou
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Dan Liu
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Fulin Chen
- College of Life Sciences, Northwest University, Xi'an, China
| | - Xiaodi Zhao
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Yuanyuan Lu
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
8
|
Fernández-Mariño I, Anfray C, Crecente-Campo J, Maeda A, Ummarino A, Teijeiro-Valiño C, Blanco-Martinez D, Mpambani F, Poul L, Devalliere J, Germain M, Correa J, Fernandez-Villamarin M, Allavena P, Fernandez-Megia E, Alonso MJ, Andón FT. Mannose-modified hyaluronic acid nanocapsules for the targeting of tumor-associated macrophages. Drug Deliv Transl Res 2023; 13:1896-1911. [PMID: 36472784 PMCID: PMC10238357 DOI: 10.1007/s13346-022-01265-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2022] [Indexed: 12/12/2022]
Abstract
Tumor-associated macrophages (TAMs), a class of immune cells that play a key role in tumor immunosuppression, are recognized as important targets to improve cancer prognosis and treatment. Consequently, the engineering of drug delivery nanocarriers that can reach TAMs has acquired special relevance. This work describes the development and biological evaluation of a panel of hyaluronic acid (HA) nanocapsules (NCs), with different compositions and prepared by different techniques, designed to target macrophages. The results showed that plain HA NCs did not significantly influence the polarization of M0 and M2-like macrophages towards an M1-like pro-inflammatory phenotype; however, the chemical functionalization of HA with mannose (HA-Man) led to a significant increase of NCs uptake by M2 macrophages in vitro and to an improved biodistribution in a MN/MNCA1 fibrosarcoma mouse model with high infiltration of TAMs. These functionalized HA-Man NCs showed a higher accumulation in the tumor compared to non-modified HA NCs. Finally, the pre-administration of the liposomal liver occupying agent Nanoprimer™ further increased the accumulation of the HA-Man NCs in the tumor. This work highlights the promise shown by the HA-Man NCs to target TAMs and thus provides new options for the development of nanomedicine and immunotherapy-based cancer treatments.
Collapse
Affiliation(s)
- Iago Fernández-Mariño
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15706, Spain
| | - Clément Anfray
- Laboratory of Cellular Immunology, IRCCS Humanitas Research Hospital, Rozzano-Milan, 20072, Italy
| | - Jose Crecente-Campo
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15706, Spain
| | - Akihiro Maeda
- Laboratory of Cellular Immunology, IRCCS Humanitas Research Hospital, Rozzano-Milan, 20072, Italy
| | - Aldo Ummarino
- Laboratory of Cellular Immunology, IRCCS Humanitas Research Hospital, Rozzano-Milan, 20072, Italy
| | - Carmen Teijeiro-Valiño
- Nanomag Laboratory, Applied Physics Department, Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Dario Blanco-Martinez
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15706, Spain
| | | | - Laurence Poul
- , Curadigm 60 rue de Wattignies, Paris, 75012, France
| | | | | | - Juan Correa
- Departamento de Química Orgánica, Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Universidade de Santiago de Compostela, Jenaro de la Fuente s/n, Santiago de Compostela, 15782, Spain
| | - Marcos Fernandez-Villamarin
- Departamento de Química Orgánica, Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Universidade de Santiago de Compostela, Jenaro de la Fuente s/n, Santiago de Compostela, 15782, Spain
| | - Paola Allavena
- Laboratory of Cellular Immunology, IRCCS Humanitas Research Hospital, Rozzano-Milan, 20072, Italy
| | - Eduardo Fernandez-Megia
- Departamento de Química Orgánica, Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS), Universidade de Santiago de Compostela, Jenaro de la Fuente s/n, Santiago de Compostela, 15782, Spain
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15706, Spain
| | - Fernando Torres Andón
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Campus Vida, Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain.
- Laboratory of Cellular Immunology, IRCCS Humanitas Research Hospital, Rozzano-Milan, 20072, Italy.
| |
Collapse
|
9
|
Zhao J, Liu L, Zhao W, Lv C, Zhang N, Jia X, Zhang Z. miR-141-3p accelerates ovarian cancer progression and promotes M2-like macrophage polarization by targeting the Keap1-Nrf2 pathway. Open Med (Wars) 2023; 18:20230729. [PMID: 37333452 PMCID: PMC10276613 DOI: 10.1515/med-2023-0729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/03/2023] [Accepted: 05/08/2023] [Indexed: 06/20/2023] Open
Abstract
The miR-141-3p has been reported to participate in regulating autophagy and tumor-stroma interactions in ovarian cancer (OC). We aim to investigate whether miR-141-3p accelerates the progression of OC and its effect on macrophage 2 polarization by targeting the Kelch-like ECH-associated protein1-Nuclear factor E2-related factor2 (Keap1-Nrf2) pathway. SKOV3 and A2780 cells were transfected with miR-141-3p inhibitor and negative control to confirm the regulation of miR-141-3p on OC development. Moreover, the growth of tumors in xenograft nude mice treated by cells transfected with miR-141-3p inhibitor was established to further testify the role of miR-141-3p in OC. The expression of miR-141-3p was higher in OC tissue compared with non-cancerous tissue. Downregulation of miR-141-3p inhibited the proliferation, migration, and invasion of ovarian cells. Furthermore, miR-141-3p inhibition also suppressed M2-like macrophage polarization and in vivo OC progression. Inhibition of miR-141-3p significantly enhanced the expression of Keap1, the target gene of miR-141-3p, and thus downregulated Nrf2, while activation of Nrf2 reversed the reduction in M2 polarization by miR-141-3p inhibitor. Collectively, miR-141-3p contributes to tumor progression, migration, and M2 polarization of OC by activating the Keap1-Nrf2 pathway. Inhibition of miR-141-3p attenuates the malignant biological behavior of ovarian cells by inactivating the Keap1-Nrf2 pathway.
Collapse
Affiliation(s)
- Jingyun Zhao
- Department of Reproductive Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Leilei Liu
- Department of Obstetrics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Wei Zhao
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Cuiting Lv
- Department of Reproductive Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Na Zhang
- Department of Reproductive Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Xinzhuan Jia
- Department of Reproductive Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Zhengmao Zhang
- Department of Gynecology, The Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| |
Collapse
|
10
|
Qiao X, Hu Z, Xiong F, Yang Y, Peng C, Wang D, Li X. Lipid metabolism reprogramming in tumor-associated macrophages and implications for therapy. Lipids Health Dis 2023; 22:45. [PMID: 37004014 PMCID: PMC10064535 DOI: 10.1186/s12944-023-01807-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 03/22/2023] [Indexed: 04/03/2023] Open
Abstract
The tumormicroenvironment (TME) plays a key role in tumor progression. Tumor-associated macrophages (TAMs), which are natural immune cells abundantin the TME, are mainly divided into the anti-tumor M1 subtype and pro-tumor M2 subtype. Due to the high plasticity of TAMs, the conversion of the M1 to M2 phenotype in hypoxic and hypoglycemic TME promotes cancer progression, which is closely related to lipid metabolism. Key factors of lipid metabolism in TAMs, including peroxisome proliferator-activated receptor and lipoxygenase, promote the formation of a tumor immunosuppressive microenvironment and facilitate immune escape. In addition, tumor cells promote lipid accumulation in TAMs, causing TAMs to polarize to the M2 phenotype. Moreover, other factors of lipid metabolism, such as abhydrolase domain containing 5 and fatty acid binding protein, have both promoting and inhibiting effects on tumor cells. Therefore, further research on lipid metabolism in tumors is still required. In addition, statins, as core drugs regulating cholesterol metabolism, can inhibit lipid rafts and adhesion of tumor cells, which can sensitize them to chemotherapeutic drugs. Clinical studies on simvastatin and lovastatin in a variety of tumors are underway. This article provides a comprehensive review of the role of lipid metabolism in TAMs in tumor progression, and provides new ideas for targeting lipid metabolism in tumor therapy.
Collapse
Affiliation(s)
- Xuehan Qiao
- Department of Medical Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhangmin Hu
- Department of Medical Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Fen Xiong
- Department of Medical Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yufei Yang
- Department of Medical Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Chen Peng
- Department of Medical Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Deqiang Wang
- Department of Medical Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Institute of Digestive Diseases, Jiangsu University, Zhenjiang, China
| | - Xiaoqin Li
- Department of Medical Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| |
Collapse
|
11
|
Dumitraş DA, Dreanca AI, Pall E, Gal AF, Rus V, Morohoschi AG, Cotul M, Nan MI, Andrei S. Inhibition of Tumor Growth and Modulation of Antioxidant Activity of Rhodoxanthin Isolated from Taxus baccata Aril against B16F10 Murine Malignant Melanoma. Antioxidants (Basel) 2022; 11:2264. [PMID: 36421450 PMCID: PMC9687082 DOI: 10.3390/antiox11112264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/10/2022] [Accepted: 11/15/2022] [Indexed: 10/29/2023] Open
Abstract
Malignant melanoma is the most aggressive type of skin cancer, and due to the numerous limitations of current treatment methods, there is an urgent need to develop novel approaches for both the prevention and treatment of malignant melanoma, with research-oriented bioactive substances representing a notable first step. The current study decided to expand on previous rhodoxanthin research by investigating the possible anti-tumor effect as well as the effect on the antioxidant status in the case of murine melanoma in an experimental model. The 21-day study was carried out on female C57BL/6J mice. On the first day of the experiment, they were subcutaneously inoculated with 106 B16F10 cells and were given rhodoxanthin orally until the end of the study. Rhodoxanthin supplementation significantly reduced tumor growth (42.18%) and weight (15.74%). Furthermore, the epidermal growth factor (EGF) activity was reduced and the concentration of 8-OHdG dropped in the treated melanoma-bearing mice compared to the untreated ones, demonstrating the role of rhodoxanthin in slowing tumor growth, one of the mechanisms being the reduction of EGF level and the decrease of DNA oxidation. The administration of rhodoxanthin determined variations in antioxidant enzymes, both at the plasma level and at the tissue level.
Collapse
Affiliation(s)
- Daria-Antonia Dumitraş
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, 400374 Cluj-Napoca, Romania
| | - Alexandra Iulia Dreanca
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, 400374 Cluj-Napoca, Romania
| | - Emoke Pall
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, 400374 Cluj-Napoca, Romania
| | - Adrian Florin Gal
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, 400374 Cluj-Napoca, Romania
| | - Vasile Rus
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, 400374 Cluj-Napoca, Romania
| | - Andreea Georgiana Morohoschi
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, 400374 Cluj-Napoca, Romania
| | - Mihaela Cotul
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, 400374 Cluj-Napoca, Romania
| | - Monica Irina Nan
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, 400374 Cluj-Napoca, Romania
| | - Sanda Andrei
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, 400374 Cluj-Napoca, Romania
| |
Collapse
|
12
|
Bartkowiak A, Nazaruk E, Gajda E, Godlewska M, Gaweł D, Jabłonowska E, Bilewicz R. Simvastatin Coadministration Modulates the Electrostatically Driven Incorporation of Doxorubicin into Model Lipid and Cell Membranes. ACS Biomater Sci Eng 2022; 8:4354-4364. [PMID: 36173110 PMCID: PMC9554873 DOI: 10.1021/acsbiomaterials.2c00724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Understanding the interactions between drugs and lipid
membranes
is a prerequisite for finding the optimal way to deliver drugs into
cells. Coadministration of statins and anticancer agents has been
reported to have a positive effect on anticancer therapy. In this
study, we elucidate the mechanism by which simvastatin (SIM) improves
the efficiency of biological membrane penetration by the chemotherapeutic
agent doxorubicin (DOX) in neutral and slightly acidic solutions.
The incorporation of DOX, SIM, or a combination of them (DOX:SIM)
into selected single-component lipid membranes, zwitterionic unsaturated
1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine
(POPC), neutral cholesterol, and negatively charged 1,2-dimyristoyl-sn-glycero-3-phospho-l-serine (DMPS) was assessed
using the Langmuir method. The penetration of neutral lipid monolayers
by the codelivery of SIM and DOX was clearly facilitated at pH 5.5,
which resembles the pH conditions of the environment of cancer cells.
This effect was ascribed to partial neutralization of the DOX positive
charge as the result of intermolecular interactions between DOX and
SIM. On the other hand, the penetration of the negatively charged
DMPS monolayer was most efficient in the case of the positively charged
DOX. The efficiency of the drug delivery to the cell membranes was
evaluated under in vitro conditions using a panel
of cancer-derived cell lines (A172, T98G, and HeLa). MTS and trypan
blue exclusion assays were performed, followed by confocal microscopy
and spheroid culture tests. Cells were exposed to either free drugs
or drugs encapsulated in lipid carriers termed cubosomes. We demonstrated
that the viability of cancer cells exposed to DOX was significantly
impaired in the presence of SIM, and this phenomenon was greatly magnified
when DOX and SIM were coencapsulated in cubosomes. Overall, our results
confirmed the utility of the DOX:SIM combination delivery, which enhances
the interactions between neutral components of cell membranes and
positively charged chemotherapeutic agents.
Collapse
Affiliation(s)
| | - Ewa Nazaruk
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02093 Warsaw, Poland
| | - Ewa Gajda
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Marlena Godlewska
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | - Damian Gaweł
- Department of Cell Biology and Immunology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | | | - Renata Bilewicz
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02093 Warsaw, Poland
| |
Collapse
|
13
|
Negrea G, Rauca VF, Meszaros MS, Patras L, Luput L, Licarete E, Toma VA, Porfire A, Muntean D, Sesarman A, Banciu M. Active Tumor-Targeting Nano-formulations Containing Simvastatin and Doxorubicin Inhibit Melanoma Growth and Angiogenesis. Front Pharmacol 2022; 13:870347. [PMID: 35450036 PMCID: PMC9016200 DOI: 10.3389/fphar.2022.870347] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 03/08/2022] [Indexed: 01/17/2023] Open
Abstract
Primary melanoma aggressiveness is determined by rapid selection and growth of cellular clones resistant to conventional treatments, resulting in metastasis and recurrence. In addition, a reprogrammed tumor-immune microenvironment supports melanoma progression and response to therapy. There is an urgent need to develop selective and specific drug delivery strategies for modulating the interaction between cancer cells and immune cells within the tumor microenvironment. This study proposes a novel combination therapy consisting of sequential administration of simvastatin incorporated in IL-13-functionalized long-circulating liposomes (IL-13-LCL-SIM) and doxorubicin encapsulated into PEG-coated extracellular vesicles (PEG-EV-DOX) to selectively target both tumor-associated macrophages and melanoma cells. To this end, IL-13 was conjugated to LCL-SIM which was obtained via the lipid film hydration method. EVs enriched from melanoma cells were passively loaded with doxorubicin. The cellular uptake of rhodamine-tagged nano-particles and the antiproliferative potential of the treatments by using the ELISA BrdU-colorimetric immunoassay were investigated in vitro. Subsequently, the therapeutic agents were administered i.v in B16.F10 melanoma-bearing mice, and tumor size was monitored during treatment. The molecular mechanisms of antitumor activity were investigated using angiogenic and inflammatory protein arrays and western blot analysis of invasion (HIF-1) and apoptosis markers (Bcl-xL and Bax). Quantification of oxidative stress marker malondialdehyde (MDA) was determined by HPLC. Immunohistochemical staining of angiogenic markers CD31 and VEGF and of pan-macrophage marker F4/80 was performed to validate our findings. The in vitro data showed that IL-13-functionalized LCL were preferentially taken up by tumor-associated macrophages and indicated that sequential administration of IL-13-LCL-SIM and PEG-EV-DOX had the strongest antiproliferative effect on tumor cells co-cultured with tumor-associated macrophages (TAMs). Accordingly, strong inhibition of tumor growth in the group treated with the sequential combination therapy was reported in vivo. Our data suggested that the antitumor action of the combined treatment was exerted through strong inhibition of several pro-angiogenic factors (VEGF, bFGF, and CD31) and oxidative stress-induced upregulation of pro-apoptotic protein Bax. This novel drug delivery strategy based on combined active targeting of both cancer cells and immune cells was able to induce a potent antitumor effect by disruption of the reciprocal interactions between TAMs and melanoma cells.
Collapse
Affiliation(s)
- Giorgiana Negrea
- Doctoral School in Integrative Biology, Faculty of Biology and Geology, "Babes-Bolyai" University, Cluj-Napoca, Romania
| | - Valentin-Florian Rauca
- Department of Molecular Biology and Biotechnology, Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, "Babes-Bolyai" University, Cluj-Napoca, Romania.,Department of Dermatology and Allergology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Marta Szilvia Meszaros
- Department of Molecular Biology and Biotechnology, Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, "Babes-Bolyai" University, Cluj-Napoca, Romania
| | - Laura Patras
- Department of Molecular Biology and Biotechnology, Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, "Babes-Bolyai" University, Cluj-Napoca, Romania
| | - Lavinia Luput
- Department of Molecular Biology and Biotechnology, Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, "Babes-Bolyai" University, Cluj-Napoca, Romania
| | - Emilia Licarete
- Department of Molecular Biology and Biotechnology, Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, "Babes-Bolyai" University, Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Vlad-Alexandru Toma
- Department of Molecular Biology and Biotechnology, Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, "Babes-Bolyai" University, Cluj-Napoca, Romania.,Department of Experimental Biology and Biochemistry, Institute of Biological Research, Branch of NIRDBS Bucharest, Cluj-Napoca, Romania
| | - Alina Porfire
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Hatieganu", Cluj-Napoca, Romania
| | - Dana Muntean
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Hatieganu", Cluj-Napoca, Romania
| | - Alina Sesarman
- Department of Molecular Biology and Biotechnology, Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, "Babes-Bolyai" University, Cluj-Napoca, Romania
| | - Manuela Banciu
- Department of Molecular Biology and Biotechnology, Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, "Babes-Bolyai" University, Cluj-Napoca, Romania
| |
Collapse
|
14
|
Zhang X, He C, Xiang G. Engineering nanomedicines to inhibit hypoxia-inducible Factor-1 for cancer therapy. Cancer Lett 2022; 530:110-127. [PMID: 35041892 DOI: 10.1016/j.canlet.2022.01.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/18/2021] [Accepted: 01/10/2022] [Indexed: 11/02/2022]
Abstract
Hypoxia-inducible factor-1 (HIF-1), an essential promoter of tumor progression, has attracted increasing attention as a therapeutic target. In addition to hypoxic cellular conditions, HIF-1 activation can be triggered by cancer treatment, which causes drug tolerance and therapeutic failure. To date, a series of effective strategies have been explored to suppress HIF-1 function, including silencing the HIF-1α gene, inhibiting HIF-1α protein translation, degrading HIF-1α protein, and inhibiting HIF-1 transcription. Furthermore, nanoparticle-based drug delivery systems have been widely developed to improve the stability and pharmacokinetics of HIF-1 inhibitors or achieve HIF-1-targeted combination therapies as a nanoplatform. In this review, we summarize the current literature on nanomedicines targeting HIF-1 to combat cancer and discuss their potential for future development.
Collapse
Affiliation(s)
- Xiaojuan Zhang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chuanchuan He
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guangya Xiang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
15
|
Rauca VF, Patras L, Luput L, Licarete E, Toma VA, Porfire A, Mot AC, Rakosy-Tican E, Sesarman A, Banciu M. Remodeling tumor microenvironment by liposomal codelivery of DMXAA and simvastatin inhibits malignant melanoma progression. Sci Rep 2021; 11:22102. [PMID: 34764332 PMCID: PMC8585864 DOI: 10.1038/s41598-021-01284-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 10/26/2021] [Indexed: 01/24/2023] Open
Abstract
Anti-angiogenic therapies for melanoma have not yet been translated into meaningful clinical benefit for patients, due to the development of drug-induced resistance in cancer cells, mainly caused by hypoxia-inducible factor 1α (HIF-1α) overexpression and enhanced oxidative stress mediated by tumor-associated macrophages (TAMs). Our previous study demonstrated synergistic antitumor actions of simvastatin (SIM) and 5,6-dimethylxanthenone-4-acetic acid (DMXAA) on an in vitro melanoma model via suppression of the aggressive phenotype of melanoma cells and inhibition of TAMs-mediated angiogenesis. Therefore, we took the advantage of long circulating liposomes (LCL) superior tumor targeting capacity to efficiently deliver SIM and DMXAA to B16.F10 melanoma in vivo, with the final aim of improving the outcome of the anti-angiogenic therapy. Thus, we assessed the effects of this novel combined tumor-targeted treatment on s.c. B16.F10 murine melanoma growth and on the production of critical markers involved in tumor development and progression. Our results showed that the combined liposomal therapy almost totally inhibited (> 90%) the growth of melanoma tumors, due to the enhancement of anti-angiogenic effects of LCL-DMXAA by LCL-SIM and simultaneous induction of a pro-apoptotic state of tumor cells in the tumor microenvironment (TME). These effects were accompanied by the partial re-education of TAMs towards an M1 phenotype and augmented by combined therapy-induced suppression of major invasion and metastasis promoters (HIF-1α, pAP-1 c-Jun, and MMPs). Thus, this novel therapy holds the potential to remodel the TME, by suppressing its most important malignant biological capabilities.
Collapse
Affiliation(s)
- Valentin-Florian Rauca
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, 5-7 Clinicilor Street, 400006, Cluj-Napoca, Romania
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, 29 Biedersteiner Street, 80802, Munich, Germany
| | - Laura Patras
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, 5-7 Clinicilor Street, 400006, Cluj-Napoca, Romania
| | - Lavinia Luput
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, 5-7 Clinicilor Street, 400006, Cluj-Napoca, Romania
| | - Emilia Licarete
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, 5-7 Clinicilor Street, 400006, Cluj-Napoca, Romania
- Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences of Babes-Bolyai University, 42 Treboniu Laurian Street, 400271, Cluj-Napoca, Romania
| | - Vlad-Alexandru Toma
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, 5-7 Clinicilor Street, 400006, Cluj-Napoca, Romania
- Department of Experimental Biology and Biochemistry, Institute of Biological Research, Branch of NIRDBS Bucharest, 48 Republicii Street, 400015, Cluj-Napoca, Romania
- Department of Molecular and Biomolecular Physics, National Institute of Research and Development for Isotopic and Molecular Technologies, 67-103 Donath Street, 400293, Cluj-Napoca, Romania
| | - Alina Porfire
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Hatieganu", 8 Babeş Street, 400012, Cluj-Napoca, Romania
| | - Augustin Catalin Mot
- Research Center for Advanced Chemical Analysis, Instrumentation and Chemometrics, Faculty of Chemistry and Chemical Engineering, Babes-Bolyai University, 11 Arany Janos Street, 400028, Cluj-Napoca, Romania
| | - Elena Rakosy-Tican
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, 5-7 Clinicilor Street, 400006, Cluj-Napoca, Romania
| | - Alina Sesarman
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, 5-7 Clinicilor Street, 400006, Cluj-Napoca, Romania.
| | - Manuela Banciu
- Department of Molecular Biology and Biotechnology, and Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, 5-7 Clinicilor Street, 400006, Cluj-Napoca, Romania
| |
Collapse
|
16
|
|
17
|
He Y, de Araújo Júnior RF, Cruz LJ, Eich C. Functionalized Nanoparticles Targeting Tumor-Associated Macrophages as Cancer Therapy. Pharmaceutics 2021; 13:1670. [PMID: 34683963 PMCID: PMC8540805 DOI: 10.3390/pharmaceutics13101670] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/02/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022] Open
Abstract
The tumor microenvironment (TME) plays a central role in regulating antitumor immune responses. As an important part of the TME, alternatively activated type 2 (M2) macrophages drive the development of primary and secondary tumors by promoting tumor cell proliferation, tumor angiogenesis, extracellular matrix remodeling and overall immunosuppression. Immunotherapy approaches targeting tumor-associated macrophages (TAMs) in order to reduce the immunosuppressive state in the TME have received great attention. Although these methods hold great potential for the treatment of several cancers, they also face some limitations, such as the fast degradation rate of drugs and drug-induced cytotoxicity of organs and tissues. Nanomedicine formulations that prevent TAM signaling and recruitment to the TME or deplete M2 TAMs to reduce tumor growth and metastasis represent encouraging novel strategies in cancer therapy. They allow the specific delivery of antitumor drugs to the tumor area, thereby reducing side effects associated with systemic application. In this review, we give an overview of TAM biology and the current state of nanomedicines that target M2 macrophages in the course of cancer immunotherapy, with a specific focus on nanoparticles (NPs). We summarize how different types of NPs target M2 TAMs, and how the physicochemical properties of NPs (size, shape, charge and targeting ligands) influence NP uptake by TAMs in vitro and in vivo in the TME. Furthermore, we provide a comparative analysis of passive and active NP-based TAM-targeting strategies and discuss their therapeutic potential.
Collapse
Affiliation(s)
- Yuanyuan He
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (Y.H.); (R.F.d.A.J.)
| | - Raimundo Fernandes de Araújo Júnior
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (Y.H.); (R.F.d.A.J.)
- Postgraduate Program in Health Science, Federal University of Rio Grande do Norte (UFRN), Natal 59064-720, Brazil
- Cancer and Inflammation Research Laboratory (LAICI), Postgraduate Program in Functional and Structural Biology, Department of Morphology, Federal University of Rio Grande do Norte (UFRN), Natal 59064-720, Brazil
- Percuros B.V., 2333 CL Leiden, The Netherlands
| | - Luis J. Cruz
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (Y.H.); (R.F.d.A.J.)
| | - Christina Eich
- Translational Nanobiomaterials and Imaging (TNI) Group, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (Y.H.); (R.F.d.A.J.)
| |
Collapse
|
18
|
Lin X, Fang Y, Jin X, Zhang M, Shi K. Modulating Repolarization of Tumor-Associated Macrophages with Targeted Therapeutic Nanoparticles as a Potential Strategy for Cancer Therapy. ACS APPLIED BIO MATERIALS 2021; 4:5871-5896. [PMID: 35006894 DOI: 10.1021/acsabm.1c00461] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There are always some components in the tumor microenvironment (TME), such as tumor-associated macrophages (TAMs), that help tumor cells escape the body's immune surveillance. Therefore, this situation can lead to tumor growth, progression, and metastasis, resulting in low response rates for cancer therapy. Macrophages play an important role with strong plasticity and functional diversity. Facing different microenvironmental stimulations, macrophages undergo a dynamic change in phenotype and function into two major macrophage subpopulations, namely classical activation/inflammation (M1) and alternative activation/regeneration (M2) type. Through various signaling pathways, macrophages polarize into complex groups, which can perform different immune functions. In this review, we emphasize the use of nanopreparations for macrophage related immunotherapy based on the pathological knowledge of TAMs phenotype. These macrophages targeted nanoparticles re-edit and re-educate macrophages by attenuating M2 macrophages and reducing aggregation to the TME, thereby relieving or alleviating immunosuppression. Among them, we describe in detail the cellular mechanisms and regulators of several major signaling pathways involved in the plasticity and polarization functions of macrophages. The advantages and challenges of those nanotherapeutics for these pathways have been elucidated, providing the basis and insights for the diagnosis and treatment strategies of various diseases centered on macrophages.
Collapse
Affiliation(s)
- Xiaojie Lin
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| | - Yan Fang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| | - Xuechao Jin
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| | - Mingming Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 117004, P. R. China
| | - Kai Shi
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, 300350 Tianjin, China
| |
Collapse
|
19
|
The potential use of simvastatin for cancer treatment: A review. Biomed Pharmacother 2021; 141:111858. [PMID: 34323700 DOI: 10.1016/j.biopha.2021.111858] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/17/2021] [Accepted: 06/28/2021] [Indexed: 12/21/2022] Open
Abstract
Statins, typically used to reduce lipid levels, have been rediscovered for exhibiting anticancer activities. Among them, especially simvastatin may influence the proliferation, migration, and survival of cancer cells. The concept of using statins to treat cancer has been adopted since the 1990s In vitro and in vivo experiments and cohort studies using statins have been carried out to demonstrate their antitumor effects (such as proliferation and migration impairment) by influencing inflammatory and oxidative stress-related tumorigenesis. Nevertheless, the biological mechanisms for these actions are not fully elucidated. In this review, we present an overview of the most important studies conducted from 2015 to date on the use of simvastatin in cancer therapy. This review brings the most recent perspectives and targets in epidemiological, in vitro, and in vivo studies, regarding the use of simvastatin alone or in combination with other drugs for the treatment of various types of cancer.
Collapse
|
20
|
Taki M, Abiko K, Ukita M, Murakami R, Yamanoi K, Yamaguchi K, Hamanishi J, Baba T, Matsumura N, Mandai M. Tumor Immune Microenvironment during Epithelial-Mesenchymal Transition. Clin Cancer Res 2021; 27:4669-4679. [PMID: 33827891 DOI: 10.1158/1078-0432.ccr-20-4459] [Citation(s) in RCA: 156] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/31/2021] [Accepted: 03/22/2021] [Indexed: 11/16/2022]
Abstract
Epithelial-mesenchymal transition (EMT) has been shown to play a critical role in tumor development from initiation to metastasis. EMT could be regarded as a continuum, with intermediate hybrid epithelial and mesenchymal phenotypes having high plasticity. Classical EMT is characterized by the phenotype change of epithelial cells to cells with mesenchymal properties, but EMT is also associated with multiple other molecular processes, including tumor immune evasion. Some previous studies have shown that EMT is associated with the cell number of immunosuppressive cells, such as myeloid-derived suppressor cells, and the expression of immune checkpoints, such as programmed cell death-ligand 1, in several cancer types. At the molecular level, EMT transcriptional factors, including Snail, Zeb1, and Twist1, produce or attract immunosuppressive cells or promote the expression of immunosuppressive checkpoint molecules via chemokine production, leading to a tumor immunosuppressive microenvironment. In turn, immunosuppressive factors induce EMT in tumor cells. This feedback loop between EMT and immunosuppression promotes tumor progression. For therapy directly targeting EMT has been challenging, the elucidation of the interactive regulation of EMT and immunosuppression is desirable for developing new therapeutic approaches in cancer. The combination of immune checkpoint inhibitors and immunotherapy targeting immunosuppressive cells could be a promising therapy for EMT.
Collapse
Affiliation(s)
- Mana Taki
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan.
| | - Kaoru Abiko
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
- Department of Obstetrics and Gynecology, National Hospital Organization Kyoto Medical Center, Fushimi-ku, Kyoto, Japan
| | - Masayo Ukita
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Ryusuke Murakami
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Koji Yamanoi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Ken Yamaguchi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Tsukasa Baba
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, Morioka, Iwate, Japan
| | - Noriomi Matsumura
- Department of Obstetrics and Gynecology, Faculty of Medicine, Kindai University, Osaka-sayama, Osaka, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| |
Collapse
|
21
|
Volovat SR, Negru S, Stolniceanu CR, Volovat C, Lungulescu C, Scripcariu D, Cobzeanu BM, Stefanescu C, Grigorescu C, Augustin I, Lupascu Ursulescu C, Volovat CC. Nanomedicine to modulate immunotherapy in cutaneous melanoma (Review). Exp Ther Med 2021; 21:535. [PMID: 33815608 PMCID: PMC8014970 DOI: 10.3892/etm.2021.9967] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer immunotherapy has shifted the paradigm in cancer treatment in recent years. Immune checkpoint blockage (ICB), the active cancer vaccination and chimeric antigen receptor (CAR) for T-cell-based adoptive cell transfer represent the main developments, achieving a surprising increased survival in patients included in clinical trials. In spite of these results, the current state-of-the-art immunotherapy has its limitations in efficacy. The existence of an interdisciplinary interface involving current knowledge in biology, immunology, bioengineering and materials science represents important progress in increasing the effectiveness of immunotherapy in cancer. Cutaneous melanoma remains a difficult cancer to treat, in which immunotherapy is a major therapeutic option. In fact, enhancing immunotherapy is possible using sophisticated biomedical nanotechnology platforms of organic or inorganic materials or engineering various immune cells to enhance the immune system. In addition, biological devices have developed, changing the approach to and treatment results in melanoma. In this review, we present different modalities to modulate the immune system, as well as opportunities and challenges in melanoma treatment.
Collapse
Affiliation(s)
- Simona Ruxandra Volovat
- Department of Medicine III-Medical Oncology-Radiotherapy, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Serban Negru
- Department of Medical Oncology, 'Victor Babes' University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Cati Raluca Stolniceanu
- Department of Biophysics and Medical Physics-Nuclear Medicine, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Constantin Volovat
- Department of Medicine III-Medical Oncology-Radiotherapy, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania.,Department of Medical Oncology, 'Euroclinic' Center of Oncology, 70010 Iasi, Romania
| | - Cristian Lungulescu
- Department of Medical Oncology, University of Medicine and Pharmacy, 200349 Craiova, Romania
| | - Dragos Scripcariu
- Department of Surgery, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Bogdan Mihail Cobzeanu
- Department of Surgery, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cipriana Stefanescu
- Department of Biophysics and Medical Physics-Nuclear Medicine, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cristina Grigorescu
- Department of Surgery, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Iolanda Augustin
- Department of Medical Oncology, 'Euroclinic' Center of Oncology, 70010 Iasi, Romania
| | - Corina Lupascu Ursulescu
- Department of Radiology, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cristian Constantin Volovat
- Department of Radiology, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iasi, Romania.,Department of Radiology, 'Sf. Spiridon' Emergency Clinic Hospital, 700111 Iasi, Romania
| |
Collapse
|
22
|
Mirzavi F, Barati M, Soleimani A, Vakili-Ghartavol R, Jaafari MR, Soukhtanloo M. A review on liposome-based therapeutic approaches against malignant melanoma. Int J Pharm 2021; 599:120413. [PMID: 33667562 DOI: 10.1016/j.ijpharm.2021.120413] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/10/2021] [Accepted: 02/17/2021] [Indexed: 01/14/2023]
Abstract
Melanoma is a highly aggressive form of skin cancer with a very poor prognosis and excessive resistance to current conventional treatments. Recently, the application of the liposomal delivery system in the management of skin melanoma has been widely investigated. Liposomal nanocarriers are biocompatible and less toxic to host cells, enabling the efficient and safe delivery of different therapeutic agents into the tumor site and further promoting their antitumor activities. Therefore, the liposomal delivery system effectively increases the success of current melanoma therapies and overcomes resistance. In this review, we present an overview of liposome-based targeted drug delivery methods and highlight recent advances towards the development of liposome-based carriers for therapeutic genes. We also discuss the new insights regarding the efficacy and clinical significance of combinatorial treatment of liposomal formulations with immunotherapy and conventional therapies in melanoma patients for a better understanding and successfully managing cancer.
Collapse
Affiliation(s)
- Farshad Mirzavi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Barati
- Department of Medical Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Anvar Soleimani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Roghayyeh Vakili-Ghartavol
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohammad Soukhtanloo
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
23
|
Boreel DF, Span PN, Heskamp S, Adema GJ, Bussink J. Targeting Oxidative Phosphorylation to Increase the Efficacy of Radio- and Immune-Combination Therapy. Clin Cancer Res 2021; 27:2970-2978. [PMID: 33419779 DOI: 10.1158/1078-0432.ccr-20-3913] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/25/2020] [Accepted: 01/06/2021] [Indexed: 11/16/2022]
Abstract
As tumors grow, they upregulate glycolytic and oxidative metabolism to support their increased and altered energetic demands. These metabolic changes have major effects on the tumor microenvironment. One of the properties leading to this aberrant metabolism is hypoxia, which occurs when tumors outgrow their often-chaotic vasculature. This scarcity of oxygen is known to induce radioresistance but can also have a disrupting effect on the antitumor immune response. Hypoxia inhibits immune effector cell function, while immune cells with a more suppressing phenotype become more active. Therefore, hypoxia strongly affects the efficacy of both radiotherapy and immunotherapy, as well as this therapy combination. Inhibition of oxidative phosphorylation (OXPHOS) is gaining interest for its ability to combat tumor hypoxia, and there are strong indications that this results in a reactivation of the immune response. This strategy decreases oxygen consumption, leading to better oxygenation of hypoxic tumor areas and eventually an increase in immunogenic cell death induced by radio-immunotherapy combinations. Promising preclinical improvements in radio- and immunotherapy efficacy have been observed by the hypoxia-reducing effect of OXPHOS inhibitors and several compounds are currently in clinical trials for their anticancer properties. Here, we will review the pharmacologic attenuation of tumor hypoxia using OXPHOS inhibitors, with emphasis on their impact on the intrinsic antitumor immune response and how this affects the efficacy of (combined) radio- and immunotherapy.
Collapse
Affiliation(s)
- Daan F Boreel
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands. .,Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Paul N Span
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Gosse J Adema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Johan Bussink
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
24
|
Shomali T, Ashrafi M. Statins, cancer, and oxidative stress. Cancer 2021. [DOI: 10.1016/b978-0-12-819547-5.00023-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
25
|
Sabra S, Agwa MM. Lactoferrin, a unique molecule with diverse therapeutical and nanotechnological applications. Int J Biol Macromol 2020; 164:1046-1060. [PMID: 32707283 PMCID: PMC7374128 DOI: 10.1016/j.ijbiomac.2020.07.167] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/10/2020] [Accepted: 07/15/2020] [Indexed: 01/25/2023]
Abstract
Lactoferrin (LF) is a naturally glycoprotein with iron-binding properties and diverse biological applications including; antiviral, anti-inflammatory, antioxidant, anti-cancer and immune stimulating effects. In addition, LF was found to be an ideal nanocarrier for some hydrophobic therapeutics because of its active targeting potential due to overexpression of its receptor on the surface of many cells. Moreover, it was proven to be a good candidate for fabrication of nanocarriers to specifically deliver drugs in case of brain tumors owing to the capability of LF to cross the blood brain barrier (BBB). Consequently, it seems to be a promising molecule with multiple applications in the field of cancer therapy and nanomedicine.
Collapse
Affiliation(s)
- Sally Sabra
- Department of Biotechnology, Institute of Graduate studies and Research, Alexandria University, Alexandria 21526, Egypt.
| | - Mona M Agwa
- Department of Chemistry of Natural and Microbial Products, Pharmaceutical and Drug Industries Research Division, National Research Centre, 33 El-Behooth St, Dokki, Giza 12311, Egypt.
| |
Collapse
|
26
|
Zhao YD, Muhetaerjiang M, An HW, Fang X, Zhao Y, Wang H. Nanomedicine enables spatiotemporally regulating macrophage-based cancer immunotherapy. Biomaterials 2020; 268:120552. [PMID: 33307365 DOI: 10.1016/j.biomaterials.2020.120552] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/12/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022]
Abstract
Cancer immunotherapy, leveraging the host's coordinated immune system to fight against tumor has been clinically validated. However, the modest response owing to the multiple ways of tumor immune evasion is one of the challenges in cancer immunotherapy. Tumor associated macrophages (TAMs), as a major component of the leukocytes infiltrating in all tumors, play crucial roles in driving cancer initiation, progress and metastasis via multiple mechanisms such as mediating chronic inflammation, promoting angiogenesis, taming protective immune responses, and supporting migration and intravasation. TAMs targeted therapeutics have achieved remarkable successes in clinical trials mostly through the use of small-molecule agents and antibodies. However, efforts for further application have met with challenges of limited efficacy and safety. Nanomaterials can provide versatile approaches to realize the superior spatiotemporal control over immunomodulation to amplify immune responses, ultimately enhancing the therapeutic benefits and reducing toxicity. Here, the potential drugs used in TAM-centered cancer treatment in clinic are summarized and the recent advances of TAMs targeted nanomedicines in this filed are highlighted. More importantly, we focus on how nanomedicine can exert their advantages in spatial and temporal control of immunomodulation.
Collapse
Affiliation(s)
- Yong-Dan Zhao
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China; Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, PR China; School of Pharmacy, Shanxi Medical University, Shanxi, 030009, PR China
| | - Mamuti Muhetaerjiang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China; GBA Research Innovation Institute for Nanotechnology, Guangdong, 510700, PR China
| | - Hong-Wei An
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China; GBA Research Innovation Institute for Nanotechnology, Guangdong, 510700, PR China
| | - Xiaohong Fang
- University of Chinese Academy of Sciences, Beijing, 100049, PR China; Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, PR China
| | - Yuliang Zhao
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, PR China; GBA Research Innovation Institute for Nanotechnology, Guangdong, 510700, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China.
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, PR China; GBA Research Innovation Institute for Nanotechnology, Guangdong, 510700, PR China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China.
| |
Collapse
|
27
|
Zhang Y, Huang J, Huang Y, Zhang S, Wu W, Long H, Duan X, Lai Y, Wu W. Tanshinone I and simvastatin inhibit melanoma tumour cell growth by regulating poly (ADP ribose) polymerase 1 expression. Mol Med Rep 2020; 23:40. [PMID: 33179075 PMCID: PMC7684874 DOI: 10.3892/mmr.2020.11678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 08/03/2020] [Indexed: 11/18/2022] Open
Abstract
Melanoma is one of the most aggressive forms of skin tumour with poor prognosis; no effective therapy has been established for melanoma at the metastatic stage. The present study aimed to investigate the role of poly (ADP ribose) polymerase (PARP) inhibitors (PARPis) and PARP1 expression in melanoma progression. In addition, whether high PARP1 expression was associated with poor overall survival in melanoma, and whether a combination effect existed between PARPis and other anti-tumour compounds (e.g., sunitinib) was analysed. The PARP1 expression was detected using western blot analysis and the proliferation of cells was detected with a colony formation assay. In addition, cell viability assays and xenograft tumor experiments were conducted. The results of the present study demonstrated that sunitinib reduced PARP1 expression and proliferation of melanoma cells. Notably, one of the PARPis, veliparib, reversed the inhibitory effect of sunitinib on PARP1 expression and proliferation, indicating that inhibition of PARP1 enzyme activity by PARPi may be different from the inhibition of PARP1 expression in melanoma cell biological function. To further confirm the relationship between PARP1 expression and tumour cell proliferation, seven compounds, including common approved drugs and natural Chinese medicine monomers, were screened, and the results demonstrated that simvastatin and tanshinone I exerted an inhibitory effect on PARP1 expression and melanoma cell proliferation, and their combination was more effective than simvastatin alone in vivo. The results indicated that simvastatin and tanshinone I inhibited melanoma and renal tumour cells by regulating PARP1 expression, and in addition to the enzyme activity of PARP1, the expression of PARP1 protein may serve a role in tumour progression.
Collapse
Affiliation(s)
- Yuyan Zhang
- Department of Pharmacy, Guangzhou Institute of Dermatology, Guangzhou, Guangdong 510095, P.R. China
| | - Jiusui Huang
- Department of Pharmacy, Guangzhou Institute of Dermatology, Guangzhou, Guangdong 510095, P.R. China
| | - Yapeng Huang
- Department of Urology, Minimally Invasive Surgery Centre, Guangzhou Urology Research Institute, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510230, P.R. China
| | - Shike Zhang
- Department of Urology, Minimally Invasive Surgery Centre, Guangzhou Urology Research Institute, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510230, P.R. China
| | - Weizhou Wu
- Department of Urology, Minimally Invasive Surgery Centre, Guangzhou Urology Research Institute, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510230, P.R. China
| | - Hui Long
- Department of Pharmacy, Guangzhou Institute of Dermatology, Guangzhou, Guangdong 510095, P.R. China
| | - Xiaolu Duan
- Department of Urology, Minimally Invasive Surgery Centre, Guangzhou Urology Research Institute, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510230, P.R. China
| | - Yongchang Lai
- Department of Urology, The Eighth Affiliated Hospital, Sun Yat‑sen University, Shenzhen, Guangdong 518033, P.R. China
| | - Wenqi Wu
- Department of Urology, Minimally Invasive Surgery Centre, Guangzhou Urology Research Institute, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510230, P.R. China
| |
Collapse
|
28
|
Yuan W, Hai B, Ren X, Zhu J, Zhang C, Guan Z, Jia J, Wang H, Cao B, Song C. Single-dose local intraosseous injection of simvastatin suppresses breast cancer with tumor vascular normalization. Transl Oncol 2020; 13:100867. [PMID: 32950929 PMCID: PMC7509234 DOI: 10.1016/j.tranon.2020.100867] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 08/05/2020] [Accepted: 08/07/2020] [Indexed: 12/23/2022] Open
Abstract
Tumor vessels play important roles in cancer development and angiogenesis has been characterized as an essential process for tumor cell tumor growth. Our previous studies found that a single-dose local intraosseous simvastatin injection rapidly and long-termly mobilized bone marrow-derived endothelial progenitor cells to peripheral blood, promoting angiogenesis and ameliorating ischemia injury. However, whether intraosseous injection of simvastatin participates in cancer progression and the role of angiogenesis enhancement in this process remain unknown. In this study, we found that intraosseous injection of simvastatin improves tumor vascular structure, along with increasing the percentage of pericyte coverage on tumor vessels, and reducing vascular permeability, tumor hypoxia and tumor necrosis. Further, we demonstrate that a single-dose local intraosseous simvastatin injection suppresses tumor growth, facilitates sensitivity of chemotherapy and prolongs survival in breast cancer-bearing mice. In addition, oral application, intravenous, subcutaneous and intraperitoneal injection of simvastatin do not show these effects. Taken together, these results demonstrate that intraosseous injection of simvastatin suppresses breast cancer with tumor vascular normalization, which might be a promising strategy for cancer treatment.
Collapse
Affiliation(s)
- Wanqiong Yuan
- Department of Orthopedics, Peking University Third Hospital, Beijing Key Laboratory of Spinal Disease, Beijing, China.
| | - Bao Hai
- Department of Orthopedics, Peking University Third Hospital, Beijing Key Laboratory of Spinal Disease, Beijing, China.
| | - Xiaoqing Ren
- Department of Pharmacy, Peking University Third Hospital, Beijing, China
| | - Junxiong Zhu
- Department of Orthopedics, Peking University Third Hospital, Beijing Key Laboratory of Spinal Disease, Beijing, China.
| | - Chenggui Zhang
- Department of Orthopedics, Peking University Third Hospital, Beijing Key Laboratory of Spinal Disease, Beijing, China.
| | - Zhiyuan Guan
- Department of Orthopedics, Peking University Third Hospital, Beijing Key Laboratory of Spinal Disease, Beijing, China.
| | - Jialin Jia
- Department of Orthopedics, Peking University Third Hospital, Beijing Key Laboratory of Spinal Disease, Beijing, China.
| | - Hong Wang
- Department of Orthopedics, Peking University Third Hospital, Beijing Key Laboratory of Spinal Disease, Beijing, China
| | - Baoshan Cao
- Department of Tumor Chemotherapy and Radiation Sickness, Peking University Third Hospital, Beijing, China
| | - Chunli Song
- Department of Orthopedics, Peking University Third Hospital, Beijing Key Laboratory of Spinal Disease, Beijing, China.
| |
Collapse
|
29
|
Preta G. New Insights Into Targeting Membrane Lipids for Cancer Therapy. Front Cell Dev Biol 2020; 8:571237. [PMID: 32984352 PMCID: PMC7492565 DOI: 10.3389/fcell.2020.571237] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/13/2020] [Indexed: 12/15/2022] Open
Abstract
Modulation of membrane lipid composition and organization is currently developing as an effective therapeutic strategy against a wide range of diseases, including cancer. This field, known as membrane-lipid therapy, has risen from new discoveries on the complex organization of lipids and between lipids and proteins in the plasma membranes. Membrane microdomains present in the membrane of all eukaryotic cells, known as lipid rafts, have been recognized as an important concentrating platform for protein receptors involved in the regulation of intracellular signaling, apoptosis, redox balance and immune response. The difference in lipid composition between the cellular membranes of healthy cells and tumor cells allows for the development of novel therapies based on targeting membrane lipids in cancer cells to increase sensitivity to chemotherapeutic agents and consequently defeat multidrug resistance. In the current manuscript strategies based on influencing cholesterol/sphingolipids content will be presented together with innovative ones, more focused in changing biophysical properties of the membrane bilayer without affecting the composition of its constituents.
Collapse
Affiliation(s)
- Giulio Preta
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
30
|
Normoxic Tumour Extracellular Vesicles Modulate the Response of Hypoxic Cancer and Stromal Cells to Doxorubicin In Vitro. Int J Mol Sci 2020; 21:ijms21175951. [PMID: 32824972 PMCID: PMC7503554 DOI: 10.3390/ijms21175951] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/03/2020] [Accepted: 08/14/2020] [Indexed: 12/19/2022] Open
Abstract
Extracellular vesicles (EV) secreted in the tumour microenvironment (TME) are emerging as major antagonists of anticancer therapies by orchestrating the therapeutic outcome through altering the behaviour of recipient cells. Recent evidence suggested that chemotherapeutic drugs could be responsible for the EV-mediated tumour-stroma crosstalk associated with cancer cell drug resistance. Here, we investigated the capacity of tumour EV (TEV) secreted by normoxic and hypoxic (1% oxygen) C26 cancer cells after doxorubicin (DOX) treatment to alter the response of naïve C26 cells and RAW 264.7 macrophages to DOX. We observed that C26 cells were less responsive to DOX treatment under normoxia compared to hypoxia, and a minimally cytotoxic DOX concentration that mounted distinct effects on cell viability was selected for TEV harvesting. Homotypic and heterotypic pretreatment of naïve hypoxic cancer and macrophage-like cells with normoxic DOX-elicited TEV rendered these cells slightly less responsive to DOX treatment. The observed effects were associated with strong hypoxia-inducible factor 1-alpha (HIF-1α) induction and B-cell lymphoma-extra-large anti-apoptotic protein (Bcl-xL)-mediated anti-apoptotic response in normoxic DOX-treated TEV donor cells, being also tightly connected to the DOX-TEV-mediated HIF-1α induction, as well as Bcl-xL levels increasing in recipient cells. Altogether, our results could open new perspectives for investigating the role of chemotherapy-elicited TEV in the colorectal cancer TME and their modulatory actions on promoting drug resistance.
Collapse
|
31
|
Licarete E, Rauca VF, Luput L, Drotar D, Stejerean I, Patras L, Dume B, Toma VA, Porfire A, Gherman C, Sesarman A, Banciu M. Overcoming Intrinsic Doxorubicin Resistance in Melanoma by Anti-Angiogenic and Anti-Metastatic Effects of Liposomal Prednisolone Phosphate on Tumor Microenvironment. Int J Mol Sci 2020; 21:ijms21082968. [PMID: 32340166 PMCID: PMC7215436 DOI: 10.3390/ijms21082968] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/13/2020] [Accepted: 04/21/2020] [Indexed: 12/31/2022] Open
Abstract
Regardless of recent progress, melanoma is very difficult to treat, mainly due to the drug resistance modulated by tumor cells as well as by the tumor microenvironment (TME). Among the immune cells recruited at the tumor site, tumor associated macrophages (TAMs) are the most abundant, promoting important tumorigenic processes: angiogenesis, inflammation and invasiveness. Furthermore, it has been shown that TAMs are involved in mediating the drug resistance of melanoma cells. Thus, in the present study, we used liposomal formulation of prednisolone disodium phosphate (LCL-PLP) to inhibit the protumor function of TAMs with the aim to sensitize the melanoma cells to the cytotoxic drug doxorubicin (DOX) to which human melanoma has intrinsic resistance. Consequently, we evaluated the in vivo effects of the concomitant administration of LCL-PLP and liposomal formulation of DOX (LCL-DOX) on B16.F10 melanoma growth and on the production of key molecular markers for tumor development. Our results demonstrated that the concomitant administration of LCL-PLP and LCL-DOX induced a strong inhibition of tumor growth, primarily by inhibiting TAMs-mediated angiogenesis as well as the tumor production of MMP-2 and AP-1. Moreover, our data suggested that the combined therapy also affected TME as the number of infiltrated macrophages in melanoma microenvironment was reduced significantly.
Collapse
Affiliation(s)
- Emilia Licarete
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania; (E.L.); (V.F.R.); (L.L.); (D.D.); (I.S.); (L.P.); (B.D.); (V.A.T.); (M.B.)
- Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, 400271 Cluj-Napoca, Romania
| | - Valentin Florian Rauca
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania; (E.L.); (V.F.R.); (L.L.); (D.D.); (I.S.); (L.P.); (B.D.); (V.A.T.); (M.B.)
- Centre of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
| | - Lavinia Luput
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania; (E.L.); (V.F.R.); (L.L.); (D.D.); (I.S.); (L.P.); (B.D.); (V.A.T.); (M.B.)
- Centre of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
| | - Denise Drotar
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania; (E.L.); (V.F.R.); (L.L.); (D.D.); (I.S.); (L.P.); (B.D.); (V.A.T.); (M.B.)
| | - Ioana Stejerean
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania; (E.L.); (V.F.R.); (L.L.); (D.D.); (I.S.); (L.P.); (B.D.); (V.A.T.); (M.B.)
| | - Laura Patras
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania; (E.L.); (V.F.R.); (L.L.); (D.D.); (I.S.); (L.P.); (B.D.); (V.A.T.); (M.B.)
- Centre of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
| | - Bogdan Dume
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania; (E.L.); (V.F.R.); (L.L.); (D.D.); (I.S.); (L.P.); (B.D.); (V.A.T.); (M.B.)
| | - Vlad Alexandru Toma
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania; (E.L.); (V.F.R.); (L.L.); (D.D.); (I.S.); (L.P.); (B.D.); (V.A.T.); (M.B.)
- Institute of Biological Research, 400015 Cluj-Napoca, Romania
- National Institute for Research and Development of Isotopic and Molecular Technologies, 400293 Cluj-Napoca, Romania
| | - Alina Porfire
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Claudia Gherman
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 400015 Cluj-Napoca, Romania;
| | - Alina Sesarman
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania; (E.L.); (V.F.R.); (L.L.); (D.D.); (I.S.); (L.P.); (B.D.); (V.A.T.); (M.B.)
- Centre of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
- Correspondence: ; Tel.: +40-264-431-691; Fax: +40-264-431-858
| | - Manuela Banciu
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania; (E.L.); (V.F.R.); (L.L.); (D.D.); (I.S.); (L.P.); (B.D.); (V.A.T.); (M.B.)
- Centre of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, 400006 Cluj-Napoca, Romania
| |
Collapse
|
32
|
Castegna A, Gissi R, Menga A, Montopoli M, Favia M, Viola A, Canton M. Pharmacological targets of metabolism in disease: Opportunities from macrophages. Pharmacol Ther 2020; 210:107521. [PMID: 32151665 DOI: 10.1016/j.pharmthera.2020.107521] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/28/2020] [Indexed: 12/12/2022]
Abstract
From advances in the knowledge of the immune system, it is emerging that the specialized functions displayed by macrophages during the course of an immune response are supported by specific and dynamically-connected metabolic programs. The study of immunometabolism is demonstrating that metabolic adaptations play a critical role in modulating inflammation and, conversely, inflammation deeply influences the acquisition of specific metabolic settings.This strict connection has been proven to be crucial for the execution of defined immune functional programs and it is now under investigation with respect to several human disorders, such as diabetes, sepsis, cancer, and autoimmunity. The abnormal remodelling of the metabolic pathways in macrophages is now emerging as both marker of disease and potential target of therapeutic intervention. By focusing on key pathological conditions, namely obesity and diabetes, rheumatoid arthritis, atherosclerosis and cancer, we will review the metabolic targets suitable for therapeutic intervention in macrophages. In addition, we will discuss the major obstacles and challenges related to the development of therapeutic strategies for a pharmacological targeting of macrophage's metabolism.
Collapse
Affiliation(s)
- Alessandra Castegna
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy; IBIOM-CNR, Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy; Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy.
| | - Rosanna Gissi
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Alessio Menga
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy; Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Monica Montopoli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy; Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
| | - Maria Favia
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Antonella Viola
- Department of Biomedical Sciences, University of Padua, Italy; Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy
| | - Marcella Canton
- Department of Biomedical Sciences, University of Padua, Italy; Fondazione Città della Speranza, Istituto di Ricerca Pediatrica, Padua, Italy.
| |
Collapse
|
33
|
Luput L, Sesarman A, Porfire A, Achim M, Muntean D, Casian T, Patras L, Rauca VF, Drotar DM, Stejerean I, Tomuta I, Vlase L, Dragos N, Toma VA, Licarete E, Banciu M. Liposomal simvastatin sensitizes C26 murine colon carcinoma to the antitumor effects of liposomal 5-fluorouracil in vivo. Cancer Sci 2020; 111:1344-1356. [PMID: 31960547 PMCID: PMC7156830 DOI: 10.1111/cas.14312] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 12/17/2019] [Accepted: 12/24/2019] [Indexed: 02/07/2023] Open
Abstract
5-Fluorouracil-based therapy remains the main approach in colorectal cancer, even though there are still some drawbacks, such as chemoresistance. In this study we combined 5-fluorouracil encapsulated in long-circulating liposomes with simvastatin, also encapsulated in long-circulating liposomes, that was previously proved to exert antitumor actions on the same tumor model. The production of angiogenic/inflammatory proteins was assessed by protein array and the production of markers for tumor aggressiveness (Bcl-2, Bax, and nuclear factor [NF]-κB) were determined by western blot analysis. Intratumor oxidative stress was evaluated through measurement of malondialdehyde level by HPLC, and through spectrophotometric analysis of catalytic activity of catalase and of total antioxidant capacity. Immunohistochemical analysis of tumors for CD31 expression was assessed. Intratumor activity of MMP-2 by gelatin zymography was also carried out. Our results revealed that combined therapies based on liposomal formulations exerted enhanced antitumor activities compared with combined treatment with free drugs. Sequential treatment with liposomal simvastatin and liposomal 5-fluorouracil showed the strongest antitumor activity in C26 colon carcinoma in vivo, mainly through inhibition of tumor angiogenesis. Important markers for cancer progression (Bcl-2, Bax, NF-κB, and intratumor antioxidants) showed that liposomal simvastatin might sensitize C26 cells to liposomal 5-fluorouracil treatment in both regimens tested. The outcome of simultaneous treatment with liposomal formulations was superior to sequential treatment with both liposomal types as the invasive capacity of C26 tumors was strongly increased after the latest treatment. The antitumor efficacy of combined therapy in C26 colon carcinoma might be linked to the restorative effects on proteins balance involved in tumor angiogenesis.
Collapse
Affiliation(s)
- Lavinia Luput
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Alina Sesarman
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Alina Porfire
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Hatieganu", Cluj-Napoca, Romania
| | - Marcela Achim
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Hatieganu", Cluj-Napoca, Romania
| | - Dana Muntean
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Hatieganu", Cluj-Napoca, Romania
| | - Tibor Casian
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Hatieganu", Cluj-Napoca, Romania
| | - Laura Patras
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Valentin Florian Rauca
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Denise Minerva Drotar
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Ioana Stejerean
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Ioan Tomuta
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Hatieganu", Cluj-Napoca, Romania
| | - Laurian Vlase
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Hatieganu", Cluj-Napoca, Romania
| | - Nicolae Dragos
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania.,Taxonomy and Ecology Department, Institute of Biological Research, Cluj-Napoca, Romania
| | - Vlad Alexandru Toma
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania.,National Institute for Research and Development of Isotopic and Molecular Technologies, Cluj-Napoca, Romania.,Department of Experimental Biology and Biochemistry, Institute of Biological Research Cluj-Napoca, branch of NIRDBS Bucharest, Cluj-Napoca, Romania
| | - Emilia Licarete
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Manuela Banciu
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, Cluj-Napoca, Romania
| |
Collapse
|
34
|
Lecocq Q, De Vlaeminck Y, Hanssens H, D'Huyvetter M, Raes G, Goyvaerts C, Keyaerts M, Devoogdt N, Breckpot K. Theranostics in immuno-oncology using nanobody derivatives. Am J Cancer Res 2019; 9:7772-7791. [PMID: 31695800 PMCID: PMC6831473 DOI: 10.7150/thno.34941] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 07/11/2019] [Indexed: 12/25/2022] Open
Abstract
Targeted therapy and immunotherapy have become mainstream in cancer treatment. However, only patient subsets benefit from these expensive therapies, and often responses are short‐lived or coincide with side effects. A growing modality in precision oncology is the development of theranostics, as this enables patient selection, treatment and monitoring. In this approach, labeled compounds and an imaging technology are used to diagnose patients and select the best treatment option, whereas for therapy, related compounds are used to target cancer cells or the tumor stroma. In this context, nanobodies and nanobody-directed therapeutics have gained interest. This interest stems from their high antigen specificity, small size, ease of labeling and engineering, allowing specific imaging and design of therapies targeting antigens on tumor cells, immune cells as well as proteins in the tumor environment. This review provides a comprehensive overview on the state-of-the-art regarding the use of nanobodies as theranostics, and their importance in the emerging field of personalized medicine.
Collapse
|
35
|
Tavakol S, Ashrafizadeh M, Deng S, Azarian M, Abdoli A, Motavaf M, Poormoghadam D, Khanbabaei H, Afshar EG, Mandegary A, Pardakhty A, Yap CT, Mohammadinejad R, Kumar AP. Autophagy Modulators: Mechanistic Aspects and Drug Delivery Systems. Biomolecules 2019; 9:E530. [PMID: 31557936 PMCID: PMC6843293 DOI: 10.3390/biom9100530] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 09/14/2019] [Accepted: 09/18/2019] [Indexed: 12/12/2022] Open
Abstract
Autophagy modulation is considered to be a promising programmed cell death mechanism to prevent and cure a great number of disorders and diseases. The crucial step in designing an effective therapeutic approach is to understand the correct and accurate causes of diseases and to understand whether autophagy plays a cytoprotective or cytotoxic/cytostatic role in the progression and prevention of disease. This knowledge will help scientists find approaches to manipulate tumor and pathologic cells in order to enhance cellular sensitivity to therapeutics and treat them. Although some conventional therapeutics suffer from poor solubility, bioavailability and controlled release mechanisms, it appears that novel nanoplatforms overcome these obstacles and have led to the design of a theranostic-controlled drug release system with high solubility and active targeting and stimuli-responsive potentials. In this review, we discuss autophagy modulators-related signaling pathways and some of the drug delivery strategies that have been applied to the field of therapeutic application of autophagy modulators. Moreover, we describe how therapeutics will target various steps of the autophagic machinery. Furthermore, nano drug delivery platforms for autophagy targeting and co-delivery of autophagy modulators with chemotherapeutics/siRNA, are also discussed.
Collapse
Affiliation(s)
- Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Milad Ashrafizadeh
- Department of basic science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| | - Shuo Deng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Maryam Azarian
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.
- Departament de Bioquímica i Biologia Molecular, Institut de Biotecnologia i Biomedicina (IBB), Universitat Autónoma de Barcelona, Barcelona, Spain.
| | - Asghar Abdoli
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran.
| | - Mahsa Motavaf
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Delaram Poormoghadam
- Department of Medical Nanotechnology, Faculty of Advanced Sciences & Technology, Pharmaceutical Sciences Branch, Islamic Azad University, (IAUPS), Tehran, Iran.
| | - Hashem Khanbabaei
- Medical Physics Department, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Elham Ghasemipour Afshar
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Ali Mandegary
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Abbas Pardakhty
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Celestial T Yap
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.
| |
Collapse
|
36
|
Zdybel M, Chodurek E, Pilawa B. Application of EPR spectroscopy to determine the influence of simvastatin concentration on free radicals in A-375 human melanoma malignum cells. Toxicol In Vitro 2019; 61:104620. [PMID: 31394162 DOI: 10.1016/j.tiv.2019.104620] [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: 12/20/2018] [Accepted: 08/02/2019] [Indexed: 10/26/2022]
Abstract
Malignant melanoma is the most aggressive and lethal form of skin cancer. Therefore the search for new methods of the treatment for melanoma is needed. In this work simvastatin was tested as the substance of potential anticancer activity. The aim of this study was to determine the effect of simvastatin in different concentration on free radicals in A-375 human melanoma malignum cells. Free radicals and growth of A-375 melanoma cells cultured without and with simvastatin were examined. Free radicals were tested by the use of electron paramagnetic resonance (EPR) spectroscopy. o-Semiquinone free radicals existed in A-375 cells. Free radicals of the control A-375 cells and the cells cultured with simvastatin were homogeneously broadened. The growth of A-375 cells did not change for concentrations of simvastatin 0.1 μM and 0.3 μM, and it slightly decreased for concentration of this substance equal 1 μM, so these concentrations of simvastatin were not preferred for therapy. For the concentrations of simvastatin higher than 1 μM the growth of A-375 cells was considerably weakened. Simvastatin in the concentrations of 0.1 μM and 1 μM decreased free radical concentrations in A-375 cells. The concentrations of simvastatin of 3 μM and 5 μM accompanied by the strong cell damage and strong formation of free radicals in them were recommended for anticancer therapy of A-375 melanoma cells.
Collapse
Affiliation(s)
- Magdalena Zdybel
- Department of Biophysics, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Jedności 8, 41-200 Sosnowiec, Poland.
| | - Ewa Chodurek
- Department of Biopharmacy, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Jedności 8, 41-200 Sosnowiec, Poland
| | - Barbara Pilawa
- Department of Biophysics, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Jedności 8, 41-200 Sosnowiec, Poland
| |
Collapse
|
37
|
Matusewicz L, Filip-Psurska B, Psurski M, Tabaczar S, Podkalicka J, Wietrzyk J, Ziółkowski P, Czogalla A, Sikorski AF. EGFR-targeted immunoliposomes as a selective delivery system of simvastatin, with potential use in treatment of triple-negative breast cancers. Int J Pharm 2019; 569:118605. [PMID: 31400433 DOI: 10.1016/j.ijpharm.2019.118605] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 08/01/2019] [Accepted: 08/06/2019] [Indexed: 11/29/2022]
Abstract
A promising strategy for treatment of EGFR-dependent tumours is EGFR signal transduction suppression via inhibition of HMG-CoA reductase using high doses of statins, popular cholesterol-lowering drugs. The main purpose of this study was to obtain targeted long circulating immunoliposomes containing simvastatin (tLCLS) with anti-EGFR antibody attached to their surface and to test whether they can be effective in treatment of TNBC. The designed tLCLS were characterized in terms of physicochemical properties and long-term stability. In vitro experiments conducted on MDA-MB-231 cells demonstrated that tLCLS induced apoptosis and are characterized by IC50 of 7.5 µM. Treatment of studied cells with tLCLS led to a decrease in membrane order and inhibited PI3K/Akt signalling. Analyses of efficacy of the tLCLS in in vivo experiments in model animals indicate that immunoliposomes were effectively delivered to tumours. Our results showed that regardless of whether tLCLS were administered before or after tumour formation, at the tested dose they inhibited tumour growth by an average of 25% in comparison to the control. However, the results were not statistically significant. The experiments described above allowed us to test the possibility of using immunoliposomes as simvastatin carriers delivering increased amounts of the drug to tumour cells.
Collapse
Affiliation(s)
- Lucyna Matusewicz
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Beata Filip-Psurska
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, ul. Weigla 12, 53-114 Wroclaw, Poland
| | - Mateusz Psurski
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, ul. Weigla 12, 53-114 Wroclaw, Poland
| | - Sabina Tabaczar
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Joanna Podkalicka
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Joanna Wietrzyk
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, ul. Weigla 12, 53-114 Wroclaw, Poland
| | - Piotr Ziółkowski
- Department of Pathology, Wroclaw Medical University, ul. K. Marcinkowskiego 1, 50-368 Wroclaw, Poland
| | - Aleksander Czogalla
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland
| | - Aleksander F Sikorski
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, ul. F. Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| |
Collapse
|
38
|
Ovais M, Guo M, Chen C. Tailoring Nanomaterials for Targeting Tumor-Associated Macrophages. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1808303. [PMID: 30883982 DOI: 10.1002/adma.201808303] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 02/07/2019] [Indexed: 05/17/2023]
Abstract
Advances in the field of nanotechnology together with an increase understanding of tumor immunology have paved the way for the development of more personalized cancer immuno-nanomedicines. Nanovehicles, due to their specific physicochemical properties, are emerging as key translational moieties in tackling tumor-promoting, M2-like tumor-associated macrophages (TAMs). Cancer immuno-nanomedicines target TAMs primarily by blocking M2-like TAM survival or affecting their signaling cascades, restricting macrophage recruitment to tumors and re-educating tumor-promoting M2-like TAMs to the tumoricidal, M1-like phenotype. Here, the TAM effector mechanisms and strategies for targeting TAMs are summarized, followed by a focus on the mechanistic considerations in the development of novel immuno-nanomedicines. Furthermore, imaging TAMs with nanoparticles so as to forecast a patient's clinical outcome, describing treatment options, and observing therapy responses is also discussed. At present, strategies that target TAMs are being investigated not only at the basic research level but also in early clinical trials. The significance of TAM-targeting biomaterials is highlighted, with the goal of facilitating future clinical translation.
Collapse
Affiliation(s)
- Muhammad Ovais
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- School of Nanoscience and Technology, College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mengyu Guo
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- School of Nanoscience and Technology, College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
- School of Nanoscience and Technology, College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
39
|
Rauca VF, Vlase L, Casian T, Sesarman A, Gheldiu AM, Mocan A, Banciu M, Toiu A. Biologically Active Ajuga Species Extracts Modulate Supportive Processes for Cancer Cell Development. Front Pharmacol 2019; 10:334. [PMID: 31024305 PMCID: PMC6460044 DOI: 10.3389/fphar.2019.00334] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 03/19/2019] [Indexed: 12/13/2022] Open
Abstract
Backround:Ajuga species have been used in traditional medicine for their diuretic, anti-inflammatory, wound-healing, and hepatoprotective properties. Purpose: The phytochemical profile and anticancer potential of three Ajuga sp. (A. genevensis, A. chamaepitys, and A. laxmannii) from Romania was investigated. Materials and Methods: The phytochemicals were extracted from the aerial parts of Ajuga sp. by using different solvents and methods. The hydroalcoholic extracts were examined for total phenolic, flavonoid and iridoid contents, and HPLC/MS was used to analyze the polyphenolic compounds and iridoids. The phytochemical profile was also evaluated by principal component analysis in connection with antitumor efficacy of extracts. The antiproliferative potential was evaluated using the ELISA BrdU-colorimetric immunoassay. Western Blot with regard to inflammatory protein NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) p65 subunit expression in cell lysates was performed. Quantification of oxidative stress marker malondialdehyde (MDA) was determined by high-performance liquid chromatography (HPLC). Enzymatic and non-enzymatic antioxidant capability was assessed by measuring catalase activity and by evaluating the total antioxidant capacity (TAC) of treated cells. Results:Ajuga laxmannii ethanol extract showed the highest total phenolic and flavonoid content, while A. genevensis ethanol extract was more abundant in iridoids. The overall cytostatic effect of the investigated plant extracts was exerted through strong inhibitory actions on NF-κB, the key molecule involved in the inflammatory response and via oxidative stress modulatory effects in both murine colon carcinoma and melanoma cell lines. Conclusion:Ajuga laxmannii showed the most significant antitumor activity and represents an important source of bioactive compounds, possibly an additional form of treatment alongside conventional anticancer drugs.
Collapse
Affiliation(s)
- Valentin-Florian Rauca
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Laurian Vlase
- Department of Pharmaceutical Technology and Biopharmacy, "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Tibor Casian
- Department of Pharmaceutical Technology and Biopharmacy, "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alina Sesarman
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Ana-Maria Gheldiu
- Department of Pharmaceutical Botany, "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Andrei Mocan
- Department of Pharmaceutical Botany, "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Manuela Banciu
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Anca Toiu
- Department of Pharmacognosy, "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
40
|
Chanier T, Chames P. Nanobody Engineering: Toward Next Generation Immunotherapies and Immunoimaging of Cancer. Antibodies (Basel) 2019; 8:E13. [PMID: 31544819 PMCID: PMC6640690 DOI: 10.3390/antib8010013] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 12/14/2022] Open
Abstract
In the last decade, cancer immunotherapies have produced impressive therapeutic results. However, the potency of immunotherapy is tightly linked to immune cell infiltration within the tumor and varies from patient to patient. Thus, it is becoming increasingly important to monitor and modulate the tumor immune infiltrate for an efficient diagnosis and therapy. Various bispecific approaches are being developed to favor immune cell infiltration through specific tumor targeting. The discovery of antibodies devoid of light chains in camelids has spurred the development of single domain antibodies (also called VHH or nanobody), allowing for an increased diversity of multispecific and/or multivalent formats of relatively small sizes endowed with high tissue penetration. The small size of nanobodies is also an asset leading to high contrasts for non-invasive imaging. The approval of the first therapeutic nanobody directed against the von Willebrand factor for the treatment of acquired thrombotic thrombocypenic purpura (Caplacizumab, Ablynx), is expected to bolster the rise of these innovative molecules. In this review, we discuss the latest advances in the development of nanobodies and nanobody-derived molecules for use in cancer immunotherapy and immunoimaging.
Collapse
Affiliation(s)
- Timothée Chanier
- Aix Marseille University, CNRS, INSERM, Institute Paoli-Calmettes, CRCM, 13009 Marseille, France.
| | - Patrick Chames
- Aix Marseille University, CNRS, INSERM, Institute Paoli-Calmettes, CRCM, 13009 Marseille, France.
| |
Collapse
|
41
|
Li X, Xiao H, Lin C, Sun W, Wu T, Wang J, Chen B, Chen X, Cheng D. Synergistic effects of liposomes encapsulating atorvastatin calcium and curcumin and targeting dysfunctional endothelial cells in reducing atherosclerosis. Int J Nanomedicine 2019; 14:649-665. [PMID: 30697048 PMCID: PMC6339643 DOI: 10.2147/ijn.s189819] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Atherosclerosis is a major cardiovascular disease that causes ischemia of the heart, brain, or extremities, and can lead to infarction. The hypolipidemic agent atorvastatin calcium (Ato) alleviates atherosclerosis by reducing plasma lipid and inflammatory factors. However, the low bioavailability of Ato limits its widespread use and clinical effectiveness. Curcumin (Cur), a natural polyphenol with antioxidation and anti-inflammation bioactivities, has potential anti-atherosclerosis activity and may reduce Ato-induced cytotoxicity. Materials and methods Liposomes modified using a targeting ligand (E-selectin-binding peptide) were prepared to co-deliver Ato and Cur to dysfunctional endothelial cells (ECs) overexpressing E-selectin. Molecules involved in the inhibition of adhesion (E-selectin and intercellular cell adhesion molecule-1 [ICAM-1]) and inflammation (IL-6 and monocyte chemotactic protein 1 [MCP-1]) in human aortic endothelial cells were evaluated using real-time quantitative PCR, flow cytometry, and immunofluorescence staining. The antiatherosclerosis effects of liposomes co-loaded with Ato and Cur in vivo were evaluated using ApoE knockout (ApoE−/−) mice. Results Targeted liposomes delivered Ato and Cur to dysfunctional ECs, resulting in synergistic suppression of adhesion molecules (E-selectin and ICAM-1) and plasma lipid levels. Moreover, this treatment reduced foam cell formation and the secretion of inflammatory factors (IL-6 and MCP-1) by blocking monocyte migration into the intima. In addition, Cur successfully reduced Ato-inducible cytotoxicity. Conclusion Both in vitro and in vivo experiments demonstrated that cell-targeted co-delivery of Ato and Cur to dysfunctional ECs drastically reduces atherosclerotic lesions with fewer side effects than either Ato or Cur alone.
Collapse
Affiliation(s)
- Xiaoxia Li
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, People's Republic of China,
| | - Hong Xiao
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, People's Republic of China,
| | - Chaowen Lin
- Department of Cardiovascular Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, People's Republic of China, .,Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Weitong Sun
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, People's Republic of China,
| | - Teng Wu
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, People's Republic of China,
| | - Jin Wang
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, People's Republic of China
| | - Bin Chen
- Department of Orthopedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Xia Chen
- Department of Cardiovascular Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, People's Republic of China,
| | - Du Cheng
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, People's Republic of China,
| |
Collapse
|
42
|
Immunoliposomes with Simvastatin as a Potential Therapeutic in Treatment of Breast Cancer Cells Overexpressing HER2-An In Vitro Study. Cancers (Basel) 2018; 10:cancers10110418. [PMID: 30388834 PMCID: PMC6266203 DOI: 10.3390/cancers10110418] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/22/2018] [Accepted: 10/30/2018] [Indexed: 12/16/2022] Open
Abstract
Lipophilic statins are promising candidates for breast cancer treatment. However, anticancer therapy requires much higher doses of statins than can be delivered orally, and such high doses are known to exert more adverse effects. The main objective of our study was to design a targeted, therapeutic liposomal carrier of simvastatin characterised by high stability and specificity towards breast cancer cells. We chose SKBR3, the cell line that showed the highest sensitivity for simvastatin and liposomal simvastatin treatment. Additionally, SKBR3 has a notably high expression level of human epidermal growth factor receptor 2 (HER2), which we used as a target for our immunoliposomes. To do so we attached humanized anti-HER2 antibody to the envelope of liposomes. We tested the stability and selectivity of the proposed formulation along with the toxicity, ability to induce apoptosis and the effect on signalling pathways involving Akt and Erk kinases. The immunoliposomal formulation of simvastatin is characterized by long-term stability, high selectivity towards HER2-overexpressing breast cancer cells, low non-specific cytotoxicity and effective inhibition of the growth of target cells, presumably by inhibition of signalling pathways and induction of apoptosis. Hence, for the first time, we propose the use of immunoliposomes with simvastatin, targeted directly towards breast cancer cells overexpressing HER2. The prepared immunoliposomes may become a proof of concept in developing new anticancer therapy.
Collapse
|
43
|
Yin W, Yu X, Kang X, Zhao Y, Zhao P, Jin H, Fu X, Wan Y, Peng C, Huang Y. Remodeling Tumor-Associated Macrophages and Neovascularization Overcomes EGFR T790M -Associated Drug Resistance by PD-L1 Nanobody-Mediated Codelivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1802372. [PMID: 30307695 DOI: 10.1002/smll.201802372] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 09/07/2018] [Indexed: 06/08/2023]
Abstract
Precision medicine has made a significant breakthrough in the past decade. The most representative success is the molecular targeting therapy of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKI) in non-small-cell lung cancer (NSCLC) with oncogenic drivers, approved by the US Food and Drug Administration (FDA) as first-line therapeutics for substituting chemotherapy. However, the rapidly developed TKI resistance invariably leads to unsustainable treatment. For example, gefitinib is the first choice for advanced NSCLC with EGFR mutation, but most patients would soon develop secondary EGFRT790M mutation and acquire gefitinib resistance. TKI resistance is a severe emergency issue to be solved in NSCLC, but there are a few investigations of nanomedicine reported to address this pressing problem. To overcome EGFRT790M -associated drug resistance, a novel delivery and therapeutic strategy is developed. A PD-L1 nanobody is identified, and first used as a targeting ligand for liposomal codelivery. It is found that simvastatin/gefitinib combination nanomedicine can remodel the tumor microenvironment (e.g., neovascularization regulation, M2-macrophage repolarization, and innate immunity), and display the effectiveness of reversing the gefitinib resistance and enhancing the EGFRT790M -mutated NSCLC treatment outcomes. The novel simvastatin-based nanomedicine provides a clinically translatable strategy for tackling the major problem in NSCLC treatment and demonstrates the promise of an old drug for new application.
Collapse
Affiliation(s)
- Weimin Yin
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, 201203, China
- Nanchang University College of Pharmacy, 461 Bayi Rd, Nanchang, 330006, China
| | - Xiaolu Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xuejia Kang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, 201203, China
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yuge Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, 201203, China
- Nanchang University College of Pharmacy, 461 Bayi Rd, Nanchang, 330006, China
| | - Pengfei Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, 201203, China
- Nanchang University College of Pharmacy, 461 Bayi Rd, Nanchang, 330006, China
| | - Hongyue Jin
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xuhong Fu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, 201203, China
- Nanchang University College of Pharmacy, 461 Bayi Rd, Nanchang, 330006, China
| | - Yakun Wan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, 201203, China
| | - Chengyuan Peng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, 201203, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
44
|
Askarizadeh A, Butler AE, Badiee A, Sahebkar A. Liposomal nanocarriers for statins: A pharmacokinetic and pharmacodynamics appraisal. J Cell Physiol 2018; 234:1219-1229. [DOI: 10.1002/jcp.27121] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/05/2018] [Indexed: 12/16/2022]
Affiliation(s)
- Anis Askarizadeh
- Nanotechnology Research Center Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
| | | | - Ali Badiee
- Nanotechnology Research Center Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
- Neurogenic Inflammation Research Center Mashhad University of Medical Sciences Mashhad Iran
- School of Pharmacy, Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
45
|
Combination therapy of simvastatin and 5, 6-dimethylxanthenone-4-acetic acid synergistically suppresses the aggressiveness of B16.F10 melanoma cells. PLoS One 2018; 13:e0202827. [PMID: 30138430 PMCID: PMC6107259 DOI: 10.1371/journal.pone.0202827] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 08/09/2018] [Indexed: 12/15/2022] Open
Abstract
The major drawback of current anti-angiogenic therapies is drug resistance, mainly caused by overexpression of the transcription factor, hypoxia-inducible factor 1α (HIF-1α) as a result of treatment-induced hypoxia, which stimulates cancer cells to develop aggressive and immunosuppressive phenotypes. Moreover, the cancer cell resistance to anti-angiogenic therapies is deeply mediated by the communication between tumor cells and tumor-associated macrophages (TAMs)-the most important microenvironmental cells for the coordination of all supportive processes in tumor development. Thus, simultaneous targeting of TAMs and cancer cells could improve the outcome of the anti-angiogenic therapies. Since our previous studies proved that simvastatin (SIM) exerts strong antiproliferative actions on B16.F10 murine melanoma cells via reduction of TAMs-mediated oxidative stress and inhibition of intratumor production of HIF-1α, we investigated whether the antitumor efficacy of the anti-angiogenic agent-5,6-dimethylxanthenone-4-acetic acid (DMXAA) could be improved by its co-administration with the lipophilic statin. Our results provide confirmatory evidence for the ability of the combined treatment to suppress the aggressive phenotype of the B16.F10 melanoma cells co-cultured with TAMs under hypoxia-mimicking conditions in vitro. Thus, proliferation and migration capacity of the melanoma cells were strongly decelerated after the co-administration of SIM and DMXAA. Moreover, our data suggested that the anti-oxidant action of the combined treatment, as a result of melanogenesis stimulation, might be the principal cause for the simultaneous suppression of key molecules involved in melanoma cell aggressiveness, present in melanoma cells (HIF-1α) as well as in TAMs (arginase-1). Finally, the concomitant suppression of these proteins might have contributed to a very strong inhibition of the angiogenic capacity of the cell co-culture microenvironment.
Collapse
|
46
|
Mishra H, Mishra PK, Ekielski A, Jaggi M, Iqbal Z, Talegaonkar S. Melanoma treatment: from conventional to nanotechnology. J Cancer Res Clin Oncol 2018; 144:2283-2302. [DOI: 10.1007/s00432-018-2726-1] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 07/30/2018] [Indexed: 11/24/2022]
|
47
|
Antiglioma via regulating oxidative stress and remodeling tumor-associated macrophage using lactoferrin-mediated biomimetic codelivery of simvastatin/fenretinide. J Control Release 2018; 287:12-23. [PMID: 30096402 DOI: 10.1016/j.jconrel.2018.08.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/22/2018] [Accepted: 08/06/2018] [Indexed: 12/13/2022]
Abstract
Effective treatment of malignant glioma still remains a formidable challenge due to lack of the effective BBB-permeable drugs and efficient brain delivery methods, and the pharmacotherapy options are very limited. Therefore, to develop an effective therapeutic strategy is a pressing need. In this work, a noncytotoxic drug combination (i.e., simvastatin and fenretinide) was revealed to be potent for treating glioma, which was co-encapsulated into a TPGS-TAT-embedded lactoferrin nanoparticle system for achieving brain-targeted biomimetic delivery via the LRP-1 receptor. It was shown that the lactoferrin nanoparticle repolarized the tumor-associated macrophages from the M2 phenotype to M1 via regulating the STAT6 pathway, as well as induced the ROS-mediated mitochondrial apoptosis by inhibiting the Ras/Raf/p-Erk pathway in the glioma cells. The antiglioma efficacy was further demonstrated in both the subcutaneous and orthotopic glioma models. The repolarization of tumor-associated macrophages not only prompted the ROS generation but also induced the innate immunity (e.g., antitumor cytokine release). This delivery and therapeutic strategy provides a novel modality for the glioma treatment.
Collapse
|
48
|
Luput L, Licarete E, Drotar DM, Nagy AL, Sesarman A, Patras L, Rauca VF, Porfire A, Muntean D, Achim M, Tomuta I, Vlase L, Catoi C, Dragos N, Banciu M. In Vivo Double Targeting of C26 Colon Carcinoma Cells and Microenvironmental Protumor Processes Using Liposomal Simvastatin. J Cancer 2018; 9:440-449. [PMID: 29344291 PMCID: PMC5771352 DOI: 10.7150/jca.21560] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 11/19/2017] [Indexed: 12/18/2022] Open
Abstract
Purpose: Besides cholesterol lowering effects, simvastatin (SIM) at very high doses possesses antitumor actions. Moreover our previous studies demonstrated that tumor-targeted delivery of SIM by using long-circulating liposomes (LCL) improved the therapeutic index of this drug in murine melanoma-bearing mice. To evaluate whether this finding can be exploited for future therapy of colorectal cancer the antitumor activity and the underlying mechanisms of long-circulating liposomal simvastatin (LCL-SIM) efficacy for inhibition of C26 murine colon carcinoma growth in vivo were investigated. Materials and Methods: To find LCL-SIM dose with the highest therapeutic index, dose-response relationship and side effects of different LCL-SIM doses were assessed in C26 colon carcinoma-bearing mice. The underlying mechanisms of LCL-SIM versus free SIM treatments were investigated with regard to their actions on C26 cell proliferation and apoptosis (via tumor tissues immunostaining for PCNA and Bax markers), tumor inflammation (via western blot analysis of NF-κΒ production), angiogenesis (using an angiogenic protein array), and oxidative stress (by HPLC assessment of malondialdehyde). Results: Our findings suggest that LCL-SIM antitumor activity on C26 colon carcinoma is a result of the tumor-targeting property of the liposome formulation, as free SIM treatment was ineffective. Moreover, LCL-SIM exerted significant antiproliferative and pro-apoptotic actions on C26 cells, notable suppressive effects on two main supportive processes for tumor development, inflammation and angiogenesis, and only slight anti-oxidant actions. Conclusion: Our data proved that LCL-SIM antitumor activity in C26 colon carcinoma was based on cytotoxic effects on these cancer cells and suppressive actions on tumor angiogenesis and inflammation.
Collapse
Affiliation(s)
- Lavinia Luput
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006, Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, 400271, Cluj-Napoca, Romania
| | - Emilia Licarete
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006, Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, 400271, Cluj-Napoca, Romania
| | - Denise Minerva Drotar
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006, Cluj-Napoca, Romania
| | - Andras-Laszlo Nagy
- Department of Veterinary Toxicology, University of Agricultural Sciences and Veterinary Medicine, 400372, Cluj Napoca, Romania
| | - Alina Sesarman
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006, Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, 400271, Cluj-Napoca, Romania
| | - Laura Patras
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006, Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, 400271, Cluj-Napoca, Romania
| | - Valentin Florian Rauca
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006, Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, 400271, Cluj-Napoca, Romania
| | - Alina Porfire
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Hatieganu", 400012, Cluj-Napoca, Romania
| | - Dana Muntean
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Hatieganu", 400012, Cluj-Napoca, Romania
| | - Marcela Achim
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Hatieganu", 400012, Cluj-Napoca, Romania
| | - Ioan Tomuta
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Hatieganu", 400012, Cluj-Napoca, Romania
| | - Laurian Vlase
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, University of Medicine and Pharmacy "Iuliu Hatieganu", 400012, Cluj-Napoca, Romania
| | - Cornel Catoi
- Department of Veterinary Toxicology, University of Agricultural Sciences and Veterinary Medicine, 400372, Cluj Napoca, Romania
| | - Nicolae Dragos
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006, Cluj-Napoca, Romania.,Taxonomy and Ecology Department, Institute of Biological Research, Cluj-Napoca, Romania
| | - Manuela Banciu
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, 400006, Cluj-Napoca, Romania.,Molecular Biology Centre, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University, 400271, Cluj-Napoca, Romania
| |
Collapse
|
49
|
Papanagnou P, Stivarou T, Papageorgiou I, Papadopoulos GE, Pappas A. Marketed drugs used for the management of hypercholesterolemia as anticancer armament. Onco Targets Ther 2017; 10:4393-4411. [PMID: 28932124 PMCID: PMC5598753 DOI: 10.2147/ott.s140483] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The design of novel pharmacologic agents as well as their approval for sale in markets all over the world is a tedious and pricey process. Inevitably, oncologic patients commonly experience unwanted effects of new anticancer drugs, while the acquisition of clinical experience for these drugs is largely based on doctor–patient partnership which is not always effective. The repositioning of marketed non-antineoplastic drugs that hopefully exhibit anticancer properties into the field of oncology is a challenging option that gains ground and attracts preclinical and clinical research in an effort to override all these hindrances and minimize the risk for reduced efficacy and/or personalized toxicity. This review aims to present the anticancer properties of drugs used for the management of hypercholesterolemia. A global view of the antitumorigenicity of all marketed antihypercholesterolemic drugs is of major importance, given that atherosclerosis, which is etiologically linked to hypercholesterolemia, is a leading worldwide cause of morbidity and mortality, while hypercholesterolemia and tumorigenesis are known to be interrelated. In vitro, in vivo and clinical literature data accumulated so far outline the mechanistic basis of the antitumor function of these agents and how they could find application at the clinical setting.
Collapse
Affiliation(s)
| | - Theodora Stivarou
- Immunology Laboratory, Immunology Department, Hellenic Pasteur Institute, Athens, Greece
| | | | | | | |
Collapse
|
50
|
Patras L, Sylvester B, Luput L, Sesarman A, Licarete E, Porfire A, Muntean D, Drotar DM, Rusu AD, Nagy AL, Catoi C, Tomuta I, Vlase L, Banciu M, Achim M. Liposomal prednisolone phosphate potentiates the antitumor activity of liposomal 5-fluorouracil in C26 murine colon carcinoma in vivo. Cancer Biol Ther 2017; 18:616-626. [PMID: 28696813 DOI: 10.1080/15384047.2017.1345392] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The antitumor efficacy of 5-fluorouracil (5-FU) in advanced colorectal cancer (CRC) is hindered not only by the low therapeutic index, but also by tumor cell resistance to this cytotoxic drug. Therefore, to enhance the 5-FU antitumor activity, the present research used a novel tumor-targeted therapy based on the co-administration of 5-FU encapsulated in long-circulating liposomes (LCL-5-FU) together with liposomal prednisolone phosphate (LCL-PLP), a formulation with known anti-angiogenic actions on C26 murine colon carcinoma cells. Thus, we assessed the in vivo effects of the combined liposomal drug therapy on C26 carcinoma growth as well as on the production of molecular markers with key roles in tumor development such as angiogenic, inflammatory, and oxidative stress molecules. To get further insight into the polarization state of tumor microenvironment after the treatment, we determined the IL-10/IL-12p70 ratio in tumors. Our results showed that combined liposomal drug therapy inhibited almost totally tumor growth and was superior as antitumor activity to both single liposomal drug therapies tested. The antitumor efficacy of the combined therapy was mainly related to the anti-angiogenic and anti-inflammatory actions on C26 carcinoma milieu, being favored by its controlling effect on intratumor oxidative stress and the skewing of polarization of tumor microenvironmental cells toward their antineoplastic phenotypes. Thus, our study unveils a promising treatment strategy for CRC that should be furthermore considered.
Collapse
Affiliation(s)
- Laura Patras
- a Department of Molecular Biology and Biotechnology , Faculty of Biology and Geology, Babes-Bolyai University , Cluj-Napoca , Romania.,b Molecular Biology Centre , Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University , Cluj-Napoca , Romania
| | - Bianca Sylvester
- c Department of Pharmaceutical Technology and Biopharmaceutics , Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy , Cluj-Napoca , Romania
| | - Lavinia Luput
- a Department of Molecular Biology and Biotechnology , Faculty of Biology and Geology, Babes-Bolyai University , Cluj-Napoca , Romania.,b Molecular Biology Centre , Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University , Cluj-Napoca , Romania
| | - Alina Sesarman
- a Department of Molecular Biology and Biotechnology , Faculty of Biology and Geology, Babes-Bolyai University , Cluj-Napoca , Romania.,b Molecular Biology Centre , Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University , Cluj-Napoca , Romania
| | - Emilia Licarete
- a Department of Molecular Biology and Biotechnology , Faculty of Biology and Geology, Babes-Bolyai University , Cluj-Napoca , Romania.,b Molecular Biology Centre , Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University , Cluj-Napoca , Romania
| | - Alina Porfire
- c Department of Pharmaceutical Technology and Biopharmaceutics , Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy , Cluj-Napoca , Romania
| | - Dana Muntean
- c Department of Pharmaceutical Technology and Biopharmaceutics , Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy , Cluj-Napoca , Romania
| | - Denise Minerva Drotar
- a Department of Molecular Biology and Biotechnology , Faculty of Biology and Geology, Babes-Bolyai University , Cluj-Napoca , Romania.,b Molecular Biology Centre , Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University , Cluj-Napoca , Romania
| | - Alexandra Doina Rusu
- a Department of Molecular Biology and Biotechnology , Faculty of Biology and Geology, Babes-Bolyai University , Cluj-Napoca , Romania.,b Molecular Biology Centre , Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University , Cluj-Napoca , Romania
| | - Andras-Laszlo Nagy
- d Department of Veterinary Toxicology , University of Agricultural Sciences and Veterinary Medicine , Cluj-Napoca , Romania
| | - Cornel Catoi
- d Department of Veterinary Toxicology , University of Agricultural Sciences and Veterinary Medicine , Cluj-Napoca , Romania
| | - Ioan Tomuta
- c Department of Pharmaceutical Technology and Biopharmaceutics , Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy , Cluj-Napoca , Romania
| | - Laurian Vlase
- c Department of Pharmaceutical Technology and Biopharmaceutics , Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy , Cluj-Napoca , Romania
| | - Manuela Banciu
- a Department of Molecular Biology and Biotechnology , Faculty of Biology and Geology, Babes-Bolyai University , Cluj-Napoca , Romania.,b Molecular Biology Centre , Institute for Interdisciplinary Research in Bio-Nano-Sciences, Babes-Bolyai University , Cluj-Napoca , Romania
| | - Marcela Achim
- c Department of Pharmaceutical Technology and Biopharmaceutics , Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy , Cluj-Napoca , Romania
| |
Collapse
|