1
|
Fukumori C, Ken Kawassaki R, Daré RG, Lopes LB. Polymer-lipid hybrid microcarriers for oral codelivery of paclitaxel and tributyrin: development, optimization, and cytotoxicity in cells and spheroids of colorectal cancer. Int J Pharm 2025; 676:125549. [PMID: 40189171 DOI: 10.1016/j.ijpharm.2025.125549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/19/2025]
Abstract
Colorectal cancer (CRC) is the third most frequent cancer worldwide. Despite advances in treatment, conventional chemotherapy suffers from severe side effects and limited drug selectivity, highlighting the importance of alternative therapies. In this study, a polymer-lipid hybrid microcarrier was developed for oral co-administration of paclitaxel (PTX) and tributyrin (TB) as a novel approach for CRC therapy. The microcarrier was designed with a pH-sensitive polymeric shell that encapsulates drug-loaded nanostructured lipid carriers (NLC); shell dissolution at intestinal pH enables localized release of the NLC. The methodological approach employed an emulsion of vegetable oil and NLC as a template for polymer deposition. Multiple parameters were optimized, including polymers ratios, NLC dilution, acid concentration, and sonication time. Spherical hybrid particles with smooth surface and mean size of 1000 nm were obtained; PTX encapsulation efficiency was 99.9 ± 0.2 %, with a production yield of 97.2 ± 0.08 %. Drug release followed the Korsmeyer-Peppas kinetic model. Cytotoxic evaluation in human colorectal adenocarcinoma HCT-116 monolayers showed that PTX encapsulation increased cytotoxicity, lowering IC50 to 83.7 nM compared to 199.5 nM for free PTX. The addition of TB further improved cytotoxicity, reducing the IC50 to 60.8 nM. A similar potentiation cytotoxicity was observed in spheroids. The microcarrier induced reductions in colony formation, alterations in cell cytoskeleton, and led to a significant reduction in P-glycoprotein expression compared to its free form, suggesting its potential to help to overcome drug resistance. These results point to the promising applicability of the hybrid microcarrier as an innovative delivery system for oral administration of cytotoxic agents.
Collapse
Affiliation(s)
- Claudio Fukumori
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Rodrigo Ken Kawassaki
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil; Department of Fundamental Chemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil
| | - Regina G Daré
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Luciana B Lopes
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
2
|
Zhang Z, Wang R, Chen L. Drug Delivery System Targeting Cancer-Associated Fibroblast for Improving Immunotherapy. Int J Nanomedicine 2025; 20:483-503. [PMID: 39816375 PMCID: PMC11734509 DOI: 10.2147/ijn.s500591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) are a heterogeneous population of non-malignant cells that play a crucial role in the tumor microenvironment, increasingly recognized as key contributors to cancer progression, metastasis, and treatment resistance. So, targeting CAFs has always been considered an important part of cancer immunotherapy. However, targeting CAFs to improve the efficacy of tumor therapy is currently a major challenge. Nanomaterials show their unique advantages in the whole process. At present, nanomaterials have achieved significant accomplishments in medical applications, particularly in the field of cancer-targeted therapy, showing enormous potential. It has been confirmed that nanomaterials can not only directly target CAFs, but also interact with the tumor microenvironment (TME) and immune cells to affect tumorigenesis. As for the cancer treatment, nanomaterials could enhance the therapeutic effect in many ways. Therefore, in this review, we first summarized the current understanding of the complex interactions between CAFs and TME, immune cells, and tumor cells. Next, we discussed common nanomaterials in modern medicine and their respective impacts on the TME, CAFs, and interactions with tumors. Finally, we focus on the application of nano drug delivery system targeting CAFs in cancer therapy.
Collapse
Affiliation(s)
- Zhongsong Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610550, People’s Republic of China
| | - Rong Wang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610550, People’s Republic of China
| | - Long Chen
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu, 610550, People’s Republic of China
| |
Collapse
|
3
|
Raaijmakers KTPM, Adema GJ, Bussink J, Ansems M. Cancer-associated fibroblasts, tumor and radiotherapy: interactions in the tumor micro-environment. J Exp Clin Cancer Res 2024; 43:323. [PMID: 39696386 DOI: 10.1186/s13046-024-03251-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) represent a group of genotypically non-malignant stromal cells in the tumor micro-environment (TME) of solid tumors that encompasses up to 80% of the tumor volume. Even though the phenotypic diversity and plasticity of CAFs complicates research, it is well-established that CAFs can affect many aspects of tumor progression, including growth, invasion and therapy resistance. Although anti-tumorigenic properties of CAFs have been reported, the majority of research demonstrates a pro-tumorigenic role for CAFs via (in)direct signaling to cancer cells, immunomodulation and extracellular matrix (ECM) remodeling. Following harsh therapeutic approaches such as radio- and/or chemotherapy, CAFs do not die but rather become senescent. Upon conversion towards senescence, many pro-tumorigenic characteristics of CAFs are preserved or even amplified. Senescent CAFs continue to promote tumor cell therapy resistance, modulate the ECM, stimulate epithelial-to-mesenchymal transition (EMT) and induce immunosuppression. Consequently, CAFs play a significant role in tumor cell survival, relapse and potentially malignant transformation of surviving cancer cells following therapy. Modulating CAF functioning in the TME therefore is a critical area of research. Proposed strategies to enhance therapeutic efficacy include reverting senescent CAFs towards a quiescent phenotype or selectively targeting (non-)senescent CAFs. In this review, we discuss CAF functioning in the TME before and during therapy, with a strong focus on radiotherapy. In the future, CAF functioning in the therapeutic TME should be taken into account when designing treatment plans and new therapeutic approaches.
Collapse
Affiliation(s)
- Kris T P M Raaijmakers
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, 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
| | - Marleen Ansems
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
4
|
Cui JY, Ma J, Gao XX, Sheng ZM, Pan ZX, Shi LH, Zhang BG. Unraveling the role of cancer-associated fibroblasts in colorectal cancer. World J Gastrointest Oncol 2024; 16:4565-4578. [PMID: 39678792 PMCID: PMC11577382 DOI: 10.4251/wjgo.v16.i12.4565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/30/2024] [Accepted: 09/19/2024] [Indexed: 11/12/2024] Open
Abstract
Within the intricate milieu of colorectal cancer (CRC) tissues, cancer-associated fibroblasts (CAFs) act as pivotal orchestrators, wielding considerable influence over tumor progression. This review endeavors to dissect the multifaceted functions of CAFs within the realm of CRC, thereby highlighting their indispensability in fostering CRC malignant microenvironment and indicating the development of CAFs-targeted therapeutic interventions. Through a comprehensive synthesis of current knowledge, this review delineates insights into CAFs-mediated modulation of cancer cell proliferation, invasiveness, immune evasion, and neovascularization, elucidating the intricate web of interactions that sustain the pro-tumor metabolism and secretion of multiple factors. Additionally, recognizing the high level of heterogeneity within CAFs is crucial, as they encompass a range of subtypes, including myofibroblastic CAFs, inflammatory CAFs, antigen-presenting CAFs, and vessel-associated CAFs. Innovatively, the symbiotic relationship between CAFs and the intestinal microbiota is explored, shedding light on a novel dimension of CRC pathogenesis. Despite remarkable progress, the orchestrated dynamic functions of CAFs remain incompletely deciphered, underscoring the need for continued research endeavors for therapeutic advancements in CRC management.
Collapse
Affiliation(s)
- Jia-Yu Cui
- Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong Province, China
| | - Jing Ma
- Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong Province, China
| | - Xin-Xin Gao
- Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong Province, China
| | - Zhi-Mei Sheng
- Affiliated Hospital of Shandong Second Medical University, Department of Pathology, Shandong Second Medical University, Weifang 261053, Shandong Province, China
| | - Zi-Xin Pan
- Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, Shandong Province, China
| | - Li-Hong Shi
- School of Rehabilitation Medicine, Shandong Second Medical University, Weifang 261053, Shandong Province, China
| | - Bao-Gang Zhang
- Department of Pathology, Shandong Second Medical University, Weifang 261053, Shandong Province, China
| |
Collapse
|
5
|
Vitale S, Calapà F, Colonna F, Luongo F, Biffoni M, De Maria R, Fiori ME. Advancements in 3D In Vitro Models for Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405084. [PMID: 38962943 PMCID: PMC11348154 DOI: 10.1002/advs.202405084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Indexed: 07/05/2024]
Abstract
The process of drug discovery and pre-clinical testing is currently inefficient, expensive, and time-consuming. Most importantly, the success rate is unsatisfactory, as only a small percentage of tested drugs are made available to oncological patients. This is largely due to the lack of reliable models that accurately predict drug efficacy and safety. Even animal models often fail to replicate human-specific pathologies and human body's complexity. These factors, along with ethical concerns regarding animal use, urge the development of suitable human-relevant, translational in vitro models.
Collapse
Affiliation(s)
- Sara Vitale
- Department of Oncology and Molecular Medicine (OMM)Istituto Superiore di SanitàViale Regina Elena 299Rome00161Italy
| | - Federica Calapà
- Dipartimento di Medicina e Chirurgia traslazionaleUniversità Cattolica del Sacro CuoreLargo F. Vito 1RomeItaly
| | - Francesca Colonna
- Department of Oncology and Molecular Medicine (OMM)Istituto Superiore di SanitàViale Regina Elena 299Rome00161Italy
| | - Francesca Luongo
- Dipartimento di Medicina e Chirurgia traslazionaleUniversità Cattolica del Sacro CuoreLargo F. Vito 1RomeItaly
| | - Mauro Biffoni
- Department of Oncology and Molecular Medicine (OMM)Istituto Superiore di SanitàViale Regina Elena 299Rome00161Italy
| | - Ruggero De Maria
- Dipartimento di Medicina e Chirurgia traslazionaleUniversità Cattolica del Sacro CuoreLargo F. Vito 1RomeItaly
- Fondazione Policlinico Universitario “A. Gemelli” – IRCCSLargo F. Vito 1RomeItaly
| | - Micol E. Fiori
- Department of Oncology and Molecular Medicine (OMM)Istituto Superiore di SanitàViale Regina Elena 299Rome00161Italy
| |
Collapse
|
6
|
Huang Q, Ge Y, He Y, Wu J, Tong Y, Shang H, Liu X, Ba X, Xia D, Peng E, Chen Z, Tang K. The Application of Nanoparticles Targeting Cancer-Associated Fibroblasts. Int J Nanomedicine 2024; 19:3333-3365. [PMID: 38617796 PMCID: PMC11012801 DOI: 10.2147/ijn.s447350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 03/23/2024] [Indexed: 04/16/2024] Open
Abstract
Cancer-associated fibroblasts (CAF) are the most abundant stromal cells in the tumor microenvironment (TME), especially in solid tumors. It has been confirmed that it can not only interact with tumor cells to promote cancer progression and metastasis, but also affect the infiltration and function of immune cells to induce chemotherapy and immunotherapy resistance. So, targeting CAF has been considered an important method in cancer treatment. The rapid development of nanotechnology provides a good perspective to improve the efficiency of targeting CAF. At present, more and more researches have focused on the application of nanoparticles (NPs) in targeting CAF. These studies explored the effects of different types of NPs on CAF and the multifunctional nanomedicines that can eliminate CAF are able to enhance the EPR effect which facilitate the anti-tumor effect of themselves. There also exist amounts of studies focusing on using NPs to inhibit the activation and function of CAF to improve the therapeutic efficacy. The application of NPs targeting CAF needs to be based on an understanding of CAF biology. Therefore, in this review, we first summarized the latest progress of CAF biology, then discussed the types of CAF-targeting NPs and the main strategies in the current. The aim is to elucidate the application of NPs in targeting CAF and provide new insights for engineering nanomedicine to enhance immune response in cancer treatment.
Collapse
Affiliation(s)
- Qiu Huang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Yue Ge
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Jian Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Yonghua Tong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Haojie Shang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Xiao Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Ding Xia
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Ejun Peng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| | - Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People’s Republic of China
| |
Collapse
|
7
|
Huang Y, Fan H, Ti H. Tumor microenvironment reprogramming by nanomedicine to enhance the effect of tumor immunotherapy. Asian J Pharm Sci 2024; 19:100902. [PMID: 38595331 PMCID: PMC11002556 DOI: 10.1016/j.ajps.2024.100902] [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: 08/28/2023] [Revised: 12/22/2023] [Accepted: 01/16/2024] [Indexed: 04/11/2024] Open
Abstract
With the rapid development of the fields of tumor biology and immunology, tumor immunotherapy has been used in clinical practice and has demonstrated significant therapeutic potential, particularly for treating tumors that do not respond to standard treatment options. Despite its advances, immunotherapy still has limitations, such as poor clinical response rates and differences in individual patient responses, largely because tumor tissues have strong immunosuppressive microenvironments. Many tumors have a tumor microenvironment (TME) that is characterized by hypoxia, low pH, and substantial numbers of immunosuppressive cells, and these are the main factors limiting the efficacy of antitumor immunotherapy. The TME is crucial to the occurrence, growth, and metastasis of tumors. Therefore, numerous studies have been devoted to improving the effects of immunotherapy by remodeling the TME. Effective regulation of the TME and reversal of immunosuppressive conditions are effective strategies for improving tumor immunotherapy. The use of multidrug combinations to improve the TME is an efficient way to enhance antitumor immune efficacy. However, the inability to effectively target drugs decreases therapeutic effects and causes toxic side effects. Nanodrug delivery carriers have the advantageous ability to enhance drug bioavailability and improve drug targeting. Importantly, they can also regulate the TME and deliver large or small therapeutic molecules to decrease the inhibitory effect of the TME on immune cells. Therefore, nanomedicine has great potential for reprogramming immunosuppressive microenvironments and represents a new immunotherapeutic strategy. Therefore, this article reviews strategies for improving the TME and summarizes research on synergistic nanomedicine approaches that enhance the efficacy of tumor immunotherapy.
Collapse
Affiliation(s)
- Yu Huang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Hui Fan
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Huihui Ti
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Province Precise Medicine Big Date of Traditional Chinese Medicine Engineering Technology Research Center, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
8
|
Freag MS, Mohammed MT, Kulkarni A, Emam HE, Maremanda KP, Elzoghby AO. Modulating tumoral exosomes and fibroblast phenotype using nanoliposomes augments cancer immunotherapy. SCIENCE ADVANCES 2024; 10:eadk3074. [PMID: 38416824 PMCID: PMC10901379 DOI: 10.1126/sciadv.adk3074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/25/2024] [Indexed: 03/01/2024]
Abstract
Cancer cells program fibroblasts into cancer associated fibroblasts (CAFs) in a two-step manner. First, cancer cells secrete exosomes to program quiescent fibroblasts into activated CAFs. Second, cancer cells maintain the CAF phenotype via activation of signal transduction pathways. We rationalized that inhibiting this two-step process can normalize CAFs into quiescent fibroblasts and augment the efficacy of immunotherapy. We show that cancer cell-targeted nanoliposomes that inhibit sequential steps of exosome biogenesis and release from lung cancer cells block the differentiation of lung fibroblasts into CAFs. In parallel, we demonstrate that CAF-targeted nanoliposomes that block two distinct nodes in fibroblast growth factor receptor (FGFR)-Wnt/β-catenin signaling pathway can reverse activate CAFs into quiescent fibroblasts. Co-administration of both nanoliposomes significantly improves the infiltration of cytotoxic T cells and enhances the antitumor efficacy of αPD-L1 in immunocompetent lung cancer-bearing mice. Simultaneously blocking the tumoral exosome-mediated activation of fibroblasts and FGFR-Wnt/β-catenin signaling constitutes a promising approach to augment immunotherapy.
Collapse
Affiliation(s)
- May S. Freag
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Investigative Toxicology, Drug Safety Research and Evaluation, Takeda Pharmaceuticals, Cambridge, MA, USA
| | - Mostafa T. Mohammed
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Anatomical and Clinical Pathology Department, Tufts Medical Center, Boston, MA, USA
| | - Arpita Kulkarni
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Hagar E. Emam
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Krishna P. Maremanda
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, USA
| | - Ahmed O. Elzoghby
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Yan L, Zheng J, Wang Q, Hao H. Role of cancer-associated fibroblasts in colorectal cancer and their potential as therapeutic targets. Biochem Biophys Res Commun 2023; 681:127-135. [PMID: 37774570 DOI: 10.1016/j.bbrc.2023.09.065] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/17/2023] [Accepted: 09/22/2023] [Indexed: 10/01/2023]
Abstract
Cancer-associated fibroblasts (CAFs) are mesenchymal cells in the tumor microenvironment (TME). CAFs are the most abundant cellular components in the TME of solid tumors. They affect the progression and course of chemotherapy and radiotherapy in various types of tumors including colorectal cancer (CRC). CAFs can promote tumor proliferation, invasion, and metastasis; protect tumor cells from immune surveillance; and resist tumor cell apoptosis caused by chemotherapy, resulting in drug resistance to chemotherapy. In recent years, researchers have become increasingly interested CAF functions and have conducted extensive research. However, compared to other types of malignancies, our understanding of the interaction between CRC cells and CAFs remains limited. Therefore, we searched the relevant literature published in the past 10 years, and reviewed the origin, biological characteristics, heterogeneity, role in the TME, and potential therapeutic targets of CAFs, to aid future research on CAFs and tumors.
Collapse
Affiliation(s)
- Liping Yan
- Department of Pathology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Jian Zheng
- Department of Pathology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, 200090, PR China
| | - Qingyu Wang
- Department of Pathology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, PR China.
| | - Hua Hao
- Department of Pathology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, 200090, PR China.
| |
Collapse
|
10
|
Wu L, Huang W, Peng K, Wang Y, Chen Q, Lu B. Enhancing the stability, BBB permeability and neuroprotective activity of verbascoside in vitro using lipid nanocapsules in combination with menthol. Food Chem 2023; 414:135682. [PMID: 36827775 DOI: 10.1016/j.foodchem.2023.135682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 01/13/2023] [Accepted: 02/09/2023] [Indexed: 02/13/2023]
Abstract
Verbascoside (VER) shows promising neuroprotective activity. However, the instability and low permeability in crossing the blood-brain barrier (BBB) greatly hinder its application. In the present study, verbascoside was encapsulated into lipid nanocapsules (LNC) by reverse micelle (RM) to increase its stability. Besides, we used VER-RM-LNC combined with an envoy drug, menthol, to improve its BBB permeability and neuroprotective activity. VER-RM-LNC was prepared by the phase inversion temperature method, resulting in an encapsulation efficiency of nearly 85 %. The formulated VER-RM-LNC was stable for 6 months at 4 °C. VER encapsulated into LNC possessed enhanced stability and a reduced release profile. Menthol increased the cellular uptake and the permeability of VER-RM-LNC in the BBB model in vitro. In addition, the improved neuroprotective activity of VER through incubation with menthol and VER-RM-LNC was verified in the neurotoxic human brain microvascular endothelial cells model induced by Aβ25-35.
Collapse
Affiliation(s)
- Lipeng Wu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China; Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China; Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311200, China
| | - Weisu Huang
- Zhejiang Institute of Economics and Trade, Hangzhou 310058, China
| | - Kejie Peng
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China; Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China; Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311200, China
| | - Yixuan Wang
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China; Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China; Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311200, China
| | - Qi Chen
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China; Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China; Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311200, China
| | - Baiyi Lu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China; Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China; Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311200, China.
| |
Collapse
|
11
|
Liang Q, Zhou XH. Role of cancer-associated fibroblasts in colorectal cancer. Shijie Huaren Xiaohua Zazhi 2023; 31:134-142. [DOI: 10.11569/wcjd.v31.i4.134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/28/2023] Open
Abstract
Colorectal cancer (CRC) is a malignancy that has a high incidence in all countries around the world. Cancer-associated fibroblasts (CAFs) are a vital component of the tumor microenvironment (TME), playing an important role in the development of CRC. CAFs can release multiple cytokines and exosomes, activating a variety of related signaling pathways and boosting the processes of the invasion, metastasis, metabolism, drug resistance, and immunosuppression in CRC. Thus, CAFs are a prognostic marker and therapeutic target for CRC. Understanding the role and mechanism of CAFs can provide new insights for the treatment of CRC.
Collapse
Affiliation(s)
- Qiao Liang
- Graduate School of Youjiang Medical College for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Xi-Han Zhou
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical College Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
12
|
Liang Q, Zhou XH, Shen GF, Zhu F, Lian HF, Li X, Zheng JY, Li JP, Deng SM, Huang R. Role of cancer-associated fibroblasts in colorectal cancer. Shijie Huaren Xiaohua Zazhi 2023; 31:129-137. [DOI: 10.11569/wcjd.v31.i4.129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/28/2023] Open
Abstract
Colorectal cancer (CRC) is a malignancy that has a high incidence in all countries around the world. Cancer-associated fibroblasts (CAFs) are a vital component of the tumor microenvironment (TME), playing an important role in the development of CRC. CAFs can release multiple cytokines and exosomes, activating a variety of related signaling pathways and boosting the processes of the invasion, metastasis, metabolism, drug resistance, and immunosuppression in CRC. Thus, CAFs are a prognostic marker and therapeutic target for CRC. Understanding the role and mechanism of CAFs can provide new insights for the treatment of CRC.
Collapse
Affiliation(s)
- Qiao Liang
- Graduate School of Youjiang Medical College for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Xi-Han Zhou
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical College Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Gao-Fei Shen
- Department of Gastroenterology, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an 710000, Shaanxi Province, China
| | - Fei Zhu
- Department of Gastroenterology, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an 710000, Shaanxi Province, China
| | - Hui-Fen Lian
- Department of Gastroenterology, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an 710000, Shaanxi Province, China
| | - Xin Li
- Department of Gastroenterology, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an 710000, Shaanxi Province, China
| | - Jun-Yi Zheng
- Department of Gastroenterology, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an 710000, Shaanxi Province, China
| | - Jin-Peng Li
- Department of Gastroenterology, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an 710000, Shaanxi Province, China
| | - Shui-Miao Deng
- Department of Gastroenterology, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an 710000, Shaanxi Province, China
| | - Rui Huang
- Department of Gastroenterology, Xi'an People's Hospital (Xi'an Fourth Hospital), Xi'an 710000, Shaanxi Province, China
| |
Collapse
|
13
|
Piorecka K, Kurjata J, Stanczyk WA. Acriflavine, an Acridine Derivative for Biomedical Application: Current State of the Art. J Med Chem 2022; 65:11415-11432. [PMID: 36018000 PMCID: PMC9469206 DOI: 10.1021/acs.jmedchem.2c00573] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Acriflavine (ACF) has been known for years as an antibacterial
drug. The identification of key oncogenic mechanisms has brought,
in recent years, a significant increase in studies on ACF as a multipurpose
drug that would improve the prognosis for cancer patients. ACF interferes
with the expression of the hypoxia inducible factor, thus acting on
metastatic niches of tumors and significantly enhancing the effects
of other anticancer therapies. It has been recognized as the most
potent HIF-1 inhibitor out of the 336 drugs approved by the FDA. This
work presents up-to-date knowledge about the mechanisms of action
of ACF and its related prodrug systems in the context of anticancer
and SARS-CoV-2 inhibitory properties. It explains the multitask nature
of this drug and suggests mechanisms of ACF’s action on the
coronavirus. Other recent reports on ACF-based systems as potential
antibacterial and antiviral drugs are also described.
Collapse
Affiliation(s)
- Kinga Piorecka
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences,Sienkiewicza 112, 90-363 Lodz, Poland
| | - Jan Kurjata
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences,Sienkiewicza 112, 90-363 Lodz, Poland
| | - Wlodzimierz A Stanczyk
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences,Sienkiewicza 112, 90-363 Lodz, Poland
| |
Collapse
|
14
|
Yu DL, Lou ZP, Ma FY, Najafi M. The interactions of paclitaxel with tumour microenvironment. Int Immunopharmacol 2022; 105:108555. [PMID: 35121223 DOI: 10.1016/j.intimp.2022.108555] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/07/2022] [Accepted: 01/17/2022] [Indexed: 12/19/2022]
Abstract
Today, it is well-known that the interactions and secretion within the tumour are crucial to consider for cancer therapy. Some novel cancer therapy modalities such as immunotherapy or tumour vaccination therapy work based on the control of interactions within the tumour microenvironment (TME). It has been revealed that anti-cancer drugs or radiotherapy can modulate some interactions in favour of cancer therapy. However, they may induce some mechanisms to increase the resistance of cancer cells to therapy. Paclitaxel is known as the first approved herbal derived chemotherapy drug. Although the main known anti-cancer effect of paclitaxel is the inhibition of the cell cycle, today, it has been well known that paclitaxel may suppress the tumour via modulating several interactions in TME. Furthermore, paclitaxel may increase the expression of some tumour resistance drivers. This review aims to discuss the interactions within TME following treatment with paclitaxel. The effects of paclitaxel on the anti-tumour immunity, immunosuppressive cells, hypoxia, and also angiogenesis will be discussed. The targeting of these interactions may be interesting to increase therapy efficiency using the combination modalities.
Collapse
Affiliation(s)
- Ding-Li Yu
- Zhuji People's Hospital of Zhejiang Province, Zhuji Affiliated Hospital of Shaoxing University, Zhuji, Zhejiang 311800, China.
| | - Zhi-Ping Lou
- Zhuji People's Hospital of Zhejiang Province, Zhuji Affiliated Hospital of Shaoxing University, Zhuji, Zhejiang 311800, China
| | - Feng-Yun Ma
- Zhuji People's Hospital of Zhejiang Province, Zhuji Affiliated Hospital of Shaoxing University, Zhuji, Zhejiang 311800, China
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran; Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
15
|
Kamali Zonouzi S, Pezeshki PS, Razi S, Rezaei N. Cancer-associated fibroblasts in colorectal cancer. Clin Transl Oncol 2021; 24:757-769. [PMID: 34839457 DOI: 10.1007/s12094-021-02734-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/06/2021] [Indexed: 12/15/2022]
Abstract
Colorectal cancer (CRC) is one of the leading causes of mortality among cancers. Many aspects of this cancer are under investigation to find established markers of diagnosis, prognosis, and also potential drug targets. In this review article, we are going to discuss the possible solution to all these aims by investigating the literature about cancer-associated fibroblasts (CAFs) involved in CRC. Moreover, we are going to review their interaction with the tumor microenvironment (TME) and vitamin D and their role in tumorigenesis and metastasis. Moreover, we are going to expand more on some markers produced by them or related to them including FAP, a-SMA, CXCL12, TGF- β, POSTN, and β1-Integrin. Some signaling pathways related to CAFs are as follows: FAK, AKT, activin A, and YAP/TAZ. Some genes related to the CAFs which are found to be possible therapeutic targets include COL3A1, JAM3, AEBP1 and, CAF-derived TGFB3, WNT2, and WNT54.
Collapse
Affiliation(s)
- S Kamali Zonouzi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - P S Pezeshki
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - S Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - N Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, 14194, Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Stockholm, Sweden.
| |
Collapse
|
16
|
[Spheroids to organoids: Solid cancer models for anticancer drug discovery]. Bull Cancer 2021; 109:49-57. [PMID: 34848046 DOI: 10.1016/j.bulcan.2021.09.019] [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: 09/06/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 11/20/2022]
Abstract
Cell culture is an important and necessary technology in oncology research. Currently, two-dimensional (2D) cell culture models are the most widely used, but they cannot reproduce the complexity and pathophysiology of tumors in vivo. This may be a major cause of the high rate of attrition of anticancer drugs entering clinical trials, the rate of new anticancer drugs entering the market being less than 5 %. One way to improve the success of new cancer drugs in the clinic is based on the use of three-dimensional (3D) cell culture models, more able to represent the complex environment and architecture of tumors. These 3D culture systems are also a powerful research tool for modeling the evolution of cancer from early stages to metastasis. Spheroids and organoids, the most adaptable models among 3D culture systems, are beginning to be used in pharmaceutical research and personalized medicine. In this article, we review the use of spheroids and organoids by highlighting their differences, discussing their impact on drug development, and looking at future challenges.
Collapse
|
17
|
Bukhari SZ, Zeth K, Iftikhar M, Rehman M, Usman Munir M, Khan WS, Ihsan A. Supramolecular lipid nanoparticles as delivery carriers for non-invasive cancer theranostics. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100067. [PMID: 34909685 PMCID: PMC8663983 DOI: 10.1016/j.crphar.2021.100067] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
Nanotheranostics is an emerging frontier of personalized medicine research particularly for cancer, which is the second leading cause of death. Supramolecular aspects in theranostics are quite allured to achieve more regulation and controlled features. Supramolecular nanotheranostics architecture is focused on engineering of modular supramolecular assemblies benefitting from their mutable and stimuli-responsive properties which confer an ultimate potential for the fabrication of unified innovative nanomedicines with controlled features. Amalgamation of supramolecular approaches to nano-based features further equip the potential of designing novel approaches to overcome limitations seen by the conventional theranostic strategies, for curing even the lethal diseases and endowing personalized therapeutics with optimistic prognosis, endorsing their clinical translation. Among many potential nanocarriers for theranostics, lipid nanoparticles (LNPs) have shown various promising advances in theranostics and their formulation can be tailored for several applications. Despite the great advancement in cancer nanotheranostics, there are still many challenges that need to be highlighted to fill the literature gap. For this purpose, herein, we have presented a systematic overview on the subject and proposed LNPs as the potential material to manage cancer via non-invasive approaches by highlighting the use of supramolecular approaches to make them robust for cancer theranostics. We have concluded the review by entailing the future perspectives of lipid nanotheranostics towards clinical translation.
Collapse
Affiliation(s)
- Syeda Zunaira Bukhari
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Kornelius Zeth
- Department of Science and Environment, Roskilde University Center, DK-4000 Roskilde, Denmark
| | - Maryam Iftikhar
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Mubashar Rehman
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, Pakistan
| | - Muhammad Usman Munir
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Aljouf, 72388, Saudi Arabia
| | - Waheed S. Khan
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Ayesha Ihsan
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| |
Collapse
|
18
|
Castro F, Leite Pereira C, Helena Macedo M, Almeida A, José Silveira M, Dias S, Patrícia Cardoso A, José Oliveira M, Sarmento B. Advances on colorectal cancer 3D models: The needed translational technology for nanomedicine screening. Adv Drug Deliv Rev 2021; 175:113824. [PMID: 34090966 DOI: 10.1016/j.addr.2021.06.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/23/2021] [Accepted: 06/01/2021] [Indexed: 02/08/2023]
Abstract
Colorectal cancer (CRC) is a heterogeneous and molecularly complex disease, associated with high mortality worldwide, exposing the urgent need for novel therapeutic approaches. Their development and translation to the clinic have been hampered, partially due to the absence of reliable cellular models that resemble key features of the human disease. While traditional 2D models are not able to provide consistent and predictive responses about the in vivo efficiency of the formulation, animal models frequently fail to recapitulate cancer progression and to reproduce adverse effects. On its turn, multicellular 3D systems, by mimicking key genetic, physical and mechanical cues of the tumor microenvironment, constitute a promising tool in cancer research. In addition, they constitute more physiological and relevant environment for anticancer drugs screening and for predicting patient's response towards personalized approaches, bridging the gap between simplified 2D models and unrepresentative animal models. In this review, we provide an overview of CRC 3D models for translational research, with focus on their potential for nanomedicines screening.
Collapse
|
19
|
El-Sheridy NA, El-Moslemany RM, Ramadan AA, Helmy MW, El-Khordagui LK. Enhancing the in vitro and in vivo activity of itraconazole against breast cancer using miltefosine-modified lipid nanocapsules. Drug Deliv 2021; 28:906-919. [PMID: 33960245 PMCID: PMC8131005 DOI: 10.1080/10717544.2021.1917728] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Itraconazole (ITC), a well-tolerated antifungal drug, exerts multiple anticancer effects which justified its preclinical and clinical investigation as potential anti-cancer agent with reduced side effects. Enhancement of ITC anti-cancer efficacy would bring valuable benefits to patients. We propose herein lipid nanocapsules (LNCs) modified with a subtherapeutic dose of miltefosine (MFS) as a membrane bioactive amphiphilic additive (M-ITC-LNC) for the development of an ITC nanoformulation with enhanced anticancer activity compared with ITC solution (ITC-sol) and unmodified ITC-LNC. Both LNC formulations showed a relatively small size (43-46 nm) and high entrapment efficiency (>97%), though ITC release was more sustained by M-ITC-LNC. Cytotoxicity studies revealed significantly greater anticancer activity and selectivity of M-ITC-LNC for MCF-7 breast cancer cells compared with ITC-sol and ITC-LNC. This trend was substantiated by in vivo findings following a 14 day-treatment of murine mammary pad Ehrlich tumors. M-ITC-LNC showed the greatest enhancement of the ITC-induced tumor growth inhibition, proliferation, and necrosis. At the molecular level, the tumor content of Gli 1, caspase-3, and vascular endothelial growth factor verified superiority of M-ITC-LNC in enhancing the ITC antiangiogenic, apoptotic, and Hedgehog pathway inhibitory effects. Finally, histopathological and biochemical analysis indicated greater reduction of ITC systemic toxicity by M-ITC-LNC. Superior performance of M-ITC-LNC was attributed to the effect of MFS on the structural and release properties of LNC coupled with its distinct bioactivities. In conclusion, MFS-modified LNC provides a simple nanoplatform integrating the potentials of LNC and MFS for enhancing the chemotherapeutic efficacy of ITC and possibly other oncology drugs.
Collapse
Affiliation(s)
- Nabila A El-Sheridy
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.,European Egyptian Pharmaceutical Industries, Alexandria, Egypt
| | - Riham M El-Moslemany
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Alyaa A Ramadan
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Maged W Helmy
- Department of Pharmacology, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Labiba K El-Khordagui
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
20
|
Joshi RS, Kanugula SS, Sudhir S, Pereira MP, Jain S, Aghi MK. The Role of Cancer-Associated Fibroblasts in Tumor Progression. Cancers (Basel) 2021; 13:cancers13061399. [PMID: 33808627 PMCID: PMC8003545 DOI: 10.3390/cancers13061399] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/10/2021] [Accepted: 03/14/2021] [Indexed: 12/15/2022] Open
Abstract
In the era of genomic medicine, cancer treatment has become more personalized as novel therapeutic targets and pathways are identified. Research over the past decade has shown the increasing importance of how the tumor microenvironment (TME) and the extracellular matrix (ECM), which is a major structural component of the TME, regulate oncogenic functions including tumor progression, metastasis, angiogenesis, therapy resistance, and immune cell modulation, amongst others. Within the TME, cancer-associated fibroblasts (CAFs) have been identified in several systemic cancers as critical regulators of the malignant cancer phenotype. This review of the literature comprehensively profiles the roles of CAFs implicated in gastrointestinal, endocrine, head and neck, skin, genitourinary, lung, and breast cancers. The ubiquitous presence of CAFs highlights their significance as modulators of cancer progression and has led to the subsequent characterization of potential therapeutic targets, which may help advance the cancer treatment paradigm to determine the next generation of cancer therapy. The aim of this review is to provide a detailed overview of the key roles that CAFs play in the scope of systemic disease, the mechanisms by which they enhance protumoral effects, and the primary CAF-related markers that may offer potential targets for novel therapeutics.
Collapse
Affiliation(s)
- Rushikesh S. Joshi
- School of Medicine, University of California, San Diego, La Jolla, CA 92092, USA;
| | | | - Sweta Sudhir
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Matheus P. Pereira
- School of Medicine, University of California, San Francisco, CA 94143, USA;
| | - Saket Jain
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA;
| | - Manish K. Aghi
- Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA;
- Correspondence: ; Tel.: +1-415-514-9820
| |
Collapse
|