1
|
Liu X, Zhang W, Wei S, Liang X, Luo B. Targeting cuproptosis with nano material: new way to enhancing the efficacy of immunotherapy in colorectal cancer. Front Pharmacol 2024; 15:1451067. [PMID: 39691393 PMCID: PMC11649426 DOI: 10.3389/fphar.2024.1451067] [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: 06/18/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024] Open
Abstract
Colorectal cancer has emerged as one of the predominant malignant tumors globally. Immunotherapy, as a novel therapeutic methodology, has opened up new possibilities for colorectal cancer patients. However, its actual clinical efficacy requires further enhancement. Copper, as an exceptionally crucial trace element, can influence various signaling pathways, gene expression, and biological metabolic processes in cells, thus playing a critical role in the pathogenesis of colorectal cancer. Recent studies have revealed that cuproptosis, a novel mode of cell death, holds promise to become a potential target to overcome resistance to colorectal cancer immunotherapy. This shows substantial potential in the combination treatment of colorectal cancer. Conveying copper into tumor cells via a nano-drug delivery system to induce cuproptosis of colorectal cancer cells could offer a potential strategy for eliminating drug-resistant colorectal cancer cells and vastly improving the efficacy of immunotherapy while ultimately destroy colorectal tumors. Moreover, combining the cuproptosis induction strategy with other anti-tumor approaches such as photothermal therapy, photodynamic therapy, and chemodynamic therapy could further enhance its therapeutic effect. This review aims to illuminate the practical significance of cuproptosis and cuproptosis-inducing nano-drugs in colorectal cancer immunotherapy, and scrutinize the current challenges and limitations of this methodology, thereby providing innovative thoughts and references for the advancement of cuproptosis-based colorectal cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Xiangdong Liu
- Department of Radiotherapy Center, Hubei Cancer Hospital, The Seventh Clinical School Affiliated of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Colorectal Cancer, Wuhan, China
- Wuhan Clinical Research Center for Colorectal Cancer, Wuhan, China
| | - Wanqiu Zhang
- Department of Radiotherapy Center, Hubei Cancer Hospital, The Seventh Clinical School Affiliated of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Colorectal Cancer, Wuhan, China
- Wuhan Clinical Research Center for Colorectal Cancer, Wuhan, China
| | - Shaozhong Wei
- Hubei Provincial Clinical Research Center for Colorectal Cancer, Wuhan, China
- Wuhan Clinical Research Center for Colorectal Cancer, Wuhan, China
- Department of Gastrointestinal Oncology Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinjun Liang
- Hubei Provincial Clinical Research Center for Colorectal Cancer, Wuhan, China
- Wuhan Clinical Research Center for Colorectal Cancer, Wuhan, China
- Department of Abdominal Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Luo
- Department of Radiotherapy Center, Hubei Cancer Hospital, The Seventh Clinical School Affiliated of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Colorectal Cancer, Wuhan, China
- Wuhan Clinical Research Center for Colorectal Cancer, Wuhan, China
| |
Collapse
|
2
|
Deng RZ, Zheng X, Lu ZL, Yuan M, Meng QC, Wu T, Tian Y. Effect of colorectal cancer stem cells on the development and metastasis of colorectal cancer. World J Gastrointest Oncol 2024; 16:4354-4368. [PMID: 39554751 PMCID: PMC11551631 DOI: 10.4251/wjgo.v16.i11.4354] [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: 07/23/2024] [Revised: 08/24/2024] [Accepted: 09/09/2024] [Indexed: 10/25/2024] Open
Abstract
The relevant mechanism of tumor-associated macrophages (TAMs) in the treatment of colorectal cancer patients with immune checkpoint inhibitors (ICIs) is discussed, and the application prospects of TAMs in reversing the treatment tolerance of ICIs are discussed to provide a reference for related studies. As a class of drugs widely used in clinical tumor immunotherapy, ICIs can act on regulatory molecules on cells that play an inhibitory role - immune checkpoints - and kill tumors in the form of an immune response by activating a variety of immune cells in the immune system. The sensitivity of patients with different types of colorectal cancer to ICI treatment varies greatly. The phenotype and function of TAMs in the colorectal cancer microenvironment are closely related to the efficacy of ICIs. ICIs can regulate the phenotypic function of TAMs, and TAMs can also affect the tolerance of colorectal cancer to ICI therapy. TAMs play an important role in ICI resistance, and making full use of this target as a therapeutic strategy is expected to improve the immunotherapy efficacy and prognosis of patients with colorectal cancer.
Collapse
Affiliation(s)
- Run-Zhi Deng
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, Fujian Province, China
| | - Xin Zheng
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, Fujian Province, China
| | - Zhong-Lei Lu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, Fujian Province, China
| | - Ming Yuan
- Department of Hepatobiliary Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
| | - Qi-Chang Meng
- Department of General Surgery, Peking University First Hospital, Beijing 100034, China
| | - Tao Wu
- Department of General Surgery, West China Hospital of Sichuan University, Chengdu 610044, Sichuan Province, China
| | - Yu Tian
- Department of Thoracic Surgery, Yancheng No. 1 People’s Hospital, Affiliated Hospital of Nanjing University Medical School, The First People’s Hospital of Yancheng, Yancheng 224000, Jiangsu Province, China
| |
Collapse
|
3
|
Gharib E, Robichaud GA. From Crypts to Cancer: A Holistic Perspective on Colorectal Carcinogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:9463. [PMID: 39273409 PMCID: PMC11395697 DOI: 10.3390/ijms25179463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Colorectal cancer (CRC) represents a significant global health burden, with high incidence and mortality rates worldwide. Recent progress in research highlights the distinct clinical and molecular characteristics of colon versus rectal cancers, underscoring tumor location's importance in treatment approaches. This article provides a comprehensive review of our current understanding of CRC epidemiology, risk factors, molecular pathogenesis, and management strategies. We also present the intricate cellular architecture of colonic crypts and their roles in intestinal homeostasis. Colorectal carcinogenesis multistep processes are also described, covering the conventional adenoma-carcinoma sequence, alternative serrated pathways, and the influential Vogelstein model, which proposes sequential APC, KRAS, and TP53 alterations as drivers. The consensus molecular CRC subtypes (CMS1-CMS4) are examined, shedding light on disease heterogeneity and personalized therapy implications.
Collapse
Affiliation(s)
- Ehsan Gharib
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| | - Gilles A Robichaud
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| |
Collapse
|
4
|
Abedizadeh R, Majidi F, Khorasani HR, Abedi H, Sabour D. Colorectal cancer: a comprehensive review of carcinogenesis, diagnosis, and novel strategies for classified treatments. Cancer Metastasis Rev 2024; 43:729-753. [PMID: 38112903 DOI: 10.1007/s10555-023-10158-3] [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] [Received: 08/08/2023] [Accepted: 11/27/2023] [Indexed: 12/21/2023]
Abstract
Colorectal cancer is the third most common and the second deadliest cancer worldwide. To date, colorectal cancer becomes one of the most important challenges of the health system in many countries. Since the clinical symptoms of this cancer appear in the final stages of the disease and there is a significant golden time between the formation of polyps and the onset of cancer, early diagnosis can play a significant role in reducing mortality. Today, in addition to colonoscopy, minimally invasive methods such as liquid biopsy have received much attention. The treatment of this complex disease has been mostly based on traditional treatments including surgery, radiotherapy, and chemotherapy; the high mortality rate indicates a lack of success for current treatment methods. Moreover, disease recurrence is another problem of traditional treatments. Recently, new approaches such as targeted therapy, immunotherapy, and nanomedicine have opened new doors for cancer treatment, some of which have already entered the market, and many methods have shown promising results in clinical trials. The success of immunotherapy in the treatment of refractory disease, the introduction of these methods into neoadjuvant therapy, and the successful results in tumor shrinkage without surgery have made immunotherapy a tough competitor for conventional treatments. It seems that the combination of those methods with such targeted therapies will go through promising changes in the future of colorectal cancer treatment.
Collapse
Affiliation(s)
- Roya Abedizadeh
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Isar 11, Babol, 47138-18983, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Bani-Hashem Square, Tehran, 16635-148, Iran
| | - Fateme Majidi
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Isar 11, Babol, 47138-18983, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Bani-Hashem Square, Tehran, 16635-148, Iran
| | - Hamid Reza Khorasani
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Isar 11, Babol, 47138-18983, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Bani-Hashem Square, Tehran, 16635-148, Iran
| | - Hassan Abedi
- Department of Internal Medicine, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran.
| | - Davood Sabour
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Isar 11, Babol, 47138-18983, Iran.
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Bani-Hashem Square, Tehran, 16635-148, Iran.
| |
Collapse
|
5
|
Shkurupii B, Zakhartseva L. PROGNOSTIC VALUE OF THE DENSITY OF TUMOR-INFILTRATING LYMPHOCYTES AND ITS ASSOCIATION WITH CLINICAL-MORPHOLOGICAL FEATURES OF COLON ADENOCARCINOMAS. Exp Oncol 2024; 46:45-52. [PMID: 38852053 DOI: 10.15407/exp-oncology.2024.01.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Indexed: 06/10/2024]
Abstract
AIM To study the prognostic value of the density of tumor-infiltrating lymphocytes (TILs) and its association with other clinical-morphological parameters in colon adenocarcinomas (CAC). MATERIALS AND METHODS 236 CAC samples were examined. TILs density was estimated as the percentage of tumor stromal area occupied by TILs. By the index of TILs density, the patients were divided into 3 groups: TILs 0-9% (n = 88); TILs 10-39% (n = 106); TILs > 40% (n = 42). Dependent on this index, their overall survival (OS) was analyzed. RESULTS Kaplan - Meier curves revealed a significant (p < 0.001) difference in the OS for patients with different TILs infiltration intensities. Multivariate Cox's proportional hazard regression model analysis has confirmed that patients with moderate TILs density (HR 0.57, 95% CI 0.34-0.96, p = 0.035) had better OS rates compared to low TILs density. TILs were associated with the stage (p < 0.001), lymph node metastasis pN (p < 0.001), distant metastasis M (p < 0.001), and the patient's outcome (p < 0.001). CONCLUSION TILs can be considered an additional prognostic tool during regular histological examination and are strongly associated with the most significant clinical-morphological features of CAC.
Collapse
Affiliation(s)
- B Shkurupii
- Bogomolets National Medical University, Kyiv, Ukraine
| | - L Zakhartseva
- Bogomolets National Medical University, Kyiv, Ukraine
- Kyiv City Oncology Hospital, Kyiv, Ukraine
| |
Collapse
|
6
|
Rahmati S, Moeinafshar A, Rezaei N. The multifaceted role of extracellular vesicles (EVs) in colorectal cancer: metastasis, immune suppression, therapy resistance, and autophagy crosstalk. J Transl Med 2024; 22:452. [PMID: 38741166 DOI: 10.1186/s12967-024-05267-8] [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: 03/19/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
Extracellular vesicles (EVs) are lipid bilayer structures released by all cells and widely distributed in all biological fluids. EVs are implicated in diverse physiopathological processes by orchestrating cell-cell communication. Colorectal cancer (CRC) is one of the most common cancers worldwide, with metastasis being the leading cause of mortality in CRC patients. EVs contribute significantly to the advancement and spread of CRC by transferring their cargo, which includes lipids, proteins, RNAs, and DNAs, to neighboring or distant cells. Besides, they can serve as non-invasive diagnostic and prognostic biomarkers for early detection of CRC or be harnessed as effective carriers for delivering therapeutic agents. Autophagy is an essential cellular process that serves to remove damaged proteins and organelles by lysosomal degradation to maintain cellular homeostasis. Autophagy and EV release are coordinately activated in tumor cells and share common factors and regulatory mechanisms. Although the significance of autophagy and EVs in cancer is well established, the exact mechanism of their interplay in tumor development is obscure. This review focuses on examining the specific functions of EVs in various aspects of CRC, including progression, metastasis, immune regulation, and therapy resistance. Further, we overview emerging discoveries relevant to autophagy and EVs crosstalk in CRC.
Collapse
Affiliation(s)
- Soheil Rahmati
- Student Research Committee, Ramsar Campus, Mazandaran University of Medical Sciences, Ramsar, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Aysan Moeinafshar
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.
- Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Sabzehei F, Taromchi AH, Ramazani A, Nedaei K, Feizi A, Arsang-Jang S, Danafar H. Cationic micelle delivery of a multi-epitope vaccine candidate derived from tumor-associated antigens, causing regression in established CT26 colorectal tumors in mice. J Biomed Mater Res A 2024; 112:733-742. [PMID: 38088136 DOI: 10.1002/jbm.a.37654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/18/2023] [Accepted: 11/27/2023] [Indexed: 03/20/2024]
Abstract
Among all the cancers, colorectal cancer (CRC) has the third mortality rank in both genders. Cancer vaccines have shown promising results in boosting patients' immune systems to fight cancer. Using the IEDB database, we predicted mouse MHC-I (H2-Ld) binding epitopes from four tumor-associated antigens (APC, KRAS, TP53, and PIK3CA) and designed a multi-epitope vaccine. We expressed the candidate vaccine and encapsulated it into the cationic micelle with polyethyleneimine conjugated to oleic acid as its building blocks. We studied tumor inhibition effect, cytokine production, and lymphocyte proliferation in the mouse CRC model after vaccination. Our finding illustrated significant tumor growth inhibition in mouse models treated with the candidate nanovaccine. Besides the significant release of IFN-γ and IL-4 by immunized mouse spleen T-lymphocytes, T-cell proliferation assay results confirmed effective immune response after the vaccination. These results demonstrate the potential therapeutic effects of nanovaccines and could be a possible approach to CRC immunotherapy.
Collapse
Affiliation(s)
- Faezeh Sabzehei
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
- Student Research Committee, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Amir Hossein Taromchi
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Ramazani
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Keivan Nedaei
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Abdolamir Feizi
- Department of Pathology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Shahram Arsang-Jang
- Department of Biostatistics and Epidemiology, School of Medicine, Znjan University of Medical Sciences, Zanjan, Iran
| | - Hossein Danafar
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
8
|
Wang G, Zhu ZM, Wang K. Identification of ROS and KEAP1-related genes and verified targets of α-hederin induce cell death for CRC. Drug Dev Res 2024; 85:e22200. [PMID: 38747107 DOI: 10.1002/ddr.22200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 04/18/2024] [Accepted: 05/06/2024] [Indexed: 06/05/2024]
Abstract
In this study, we analyzed and verified differentially expressed genes (DEGs) in ROS and KEAP1 crosstalk in oncogenic signatures using GEO data sets (GSE4107 and GSE41328). Multiple pathway enrichment analyses were finished based on DEGs. The genetic signature for colorectal adenocarcinoma (COAD) was identified by using the Cox regression analysis. Kaplan-Meier survival and receiver operating characteristic curve analysis were used to explore the prognosis value of specific genes in COAD. The potential immune signatures and drug sensitivity prediction were also analyzed. Promising small-molecule agents were identified and predicted targets of α-hederin in SuperPred were validated by molecular docking. Also, expression levels of genes and Western blot analysis were conducted. In total, 48 genes were identified as DEGs, and the hub genes such as COL1A1, CXCL12, COL1A2, FN1, CAV1, TIMP3, and IGFBP7 were identified. The ROS and KEAP1-associated gene signatures comprised of hub key genes were developed for predicting the prognosis and evaluating the immune cell responses and immune infiltration in COAD. α-hederin, a potential anti-colorectal cancer (CRC) agent, was found to enhance the sensitivity of HCT116 cells, regulate CAV1 and COL1A1, and decrease KEAP1, Nrf2, and HO-1 expression significantly. KEAP1-related genes could be an essential mediator of ROS in CRC, and KEAP1-associated genes were effective in predicting prognosis and evaluating individualized CRC treatment. Therefore, α-hederin may be an effective chemosensitizer for CRC treatments in clinical settings.
Collapse
Affiliation(s)
- Gang Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Zhi-Min Zhu
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Kun Wang
- Department of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
9
|
Zhu S, Li X, Luo Z, Ding M, Shi S, Zhang T. Combined immunochemotherapy achieving targeted co-delivery of chlorogenic acid and doxorubicin by sialic acid-modified liposomes enhances anti-cancer efficacy. Drug Deliv Transl Res 2024; 14:718-729. [PMID: 37679600 DOI: 10.1007/s13346-023-01426-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2023] [Indexed: 09/09/2023]
Abstract
Malignant melanoma is a high-grade aggressive skin tumor with an increasing incidence and mortality rates worldwide. Chemotherapeutic drugs such as doxorubicin have limited efficacy against melanoma due to their poor sensitivity, severe side effects, and drug resistance. Recent studies have shown that combinations of immunotherapy and chemotherapy have a synergistic effect in enhancing the anti-tumor effect. Here, we have developed liposomes co-loaded with chlorogenic acid (CA) and doxorubicin (DOX), modified with sialic acid-octadecylamine conjugate (SA-ODA), designated CA-DOX-SAL, that facilitate drug delivery by recognizing Siglec-1 receptor on TAMs. The physicochemical studies revealed the particle size and zeta potential of CA-DOX-SAL as 128.3 ± 0.8 nm and - 4.33 ± 0.50 mV, respectively. In vitro, CA-DOX-SAL demonstrated robust cellular uptake through SA receptor-mediated tumor-associated macrophages (TAM) targeting and exerted greater cytotoxicity on tumor cells. In vivo, targeted liposomes were found to accumulate in the tumor area, leading to an improvement in anti-tumor efficacy. In addition, CA-DOX-SAL effectively inhibited B16F10 melanoma tumor growth by stimulating the transition from tumor-promoting M2-type to anti-tumor M1-type and directly killing tumor cells. Overall, the co-delivery of immunomodulatory CA and chemotherapeutic DOX presents a promising therapeutic strategy to enhance clinical outcomes in the treatment of melanoma.
Collapse
Affiliation(s)
- Shunyao Zhu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xixi Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Ziyi Luo
- Department of Pharmacy, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310018, China
| | - Meihong Ding
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Senlin Shi
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Ting Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
10
|
Cao Y, Wo M, Xu C, Fei X, Jin J, Shan Z. An AMPK agonist suppresses the progress of colorectal cancer by regulating the polarization of TAM to M1 through inhibition of HIF-1α and mTOR signal pathway. J Cancer Res Ther 2023; 19:1560-1567. [PMID: 38156922 DOI: 10.4103/jcrt.jcrt_2670_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 07/17/2023] [Indexed: 01/03/2024]
Abstract
OBJECTIVE This study aimed to evaluate the impact of an adenosine monophosphate-activated protein kinase (AMPK) agonist, metformin (MET), on the antitumor effects of macrophages and to determine the underlying mechanism involved in the process. MATERIALS AND METHODS M0 macrophages were derived from phorbol-12-myristate-13-acetate-stimulated THP-1 cells. RESULTS The levels of tumor necrosis factor-alpha (TNF-α) and human leukocyte antigen-DR (HLA-DR) were decreased in macrophages incubated with HCT116 cells, whereas those of arginase-1 (Arg-1), CD163, and CD206 were elevated; these effects were reversed by MET. The transfection of small interfering (si) RNA abrogated the influence of MET on the expression of the M1/M2 macrophage biomarkers. MET significantly suppressed the proliferation and migration abilities of HCT116 cells incubated with M0 macrophages; these actions were reversed by siRNA transfection against AMPK. The hypoxia-inducible factor 1-alpha (HIF-1α), phosphorylated protein kinase B (p-AKT), and phosphorylated mammalian target of rapamycin (p-mTOR) levels were reduced by the introduction of MET and promoted by siRNA transfection against AMPK. In addition, the levels of HIF-1α, p-AKT, and p-mTOR suppressed by MET were markedly increased following the transfection of siRNA against AMPK. CONCLUSION These findings indicate that MET can repress the progression of colorectal cancer by transforming tumor-associated macrophages to the M1phenotype via inhibition of the HIF-1α and mTOR signaling pathways.
Collapse
Affiliation(s)
- Yuanyuan Cao
- Department of Laboratory Medicine, Hangzhou Cancer Hospital, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Shangcheng, China
| | - Mingyi Wo
- Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Zhejiang Center for Clinical Laboratory, Gongshu, China
| | - Chan Xu
- Department of Laboratory Medicine, Affiliated Third Hospital of Zhejiang Traditional Chinese Medicine University, Xihu, Hangzhou, Zhejiang, China
| | - Xianming Fei
- Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Zhejiang Center for Clinical Laboratory, Gongshu, China
| | - Juan Jin
- Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Zhejiang Center for Clinical Laboratory, Gongshu, China
| | - Zhiming Shan
- Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Zhejiang Center for Clinical Laboratory, Gongshu, China
| |
Collapse
|
11
|
Yang X, Luo Y, Li M, Jin Z, Chen G, Gan C. Long non-coding RNA NBR2 suppresses the progression of colorectal cancer by downregulating miR-19a to regulate M2 macrophage polarization. CHINESE J PHYSIOL 2023; 66:546-557. [PMID: 38149567 DOI: 10.4103/cjop.cjop-d-23-00064] [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] [Indexed: 12/28/2023] Open
Abstract
Colorectal cancer (CRC) is a malignant tumor of the gastrointestinal tract that significantly impacts the health of patients and lacks promising methods of diagnosis. Tumor-associated macrophages (TAMs) are involved in CRC progression, and their function is regulated by long non-coding RNAs (lncRNAs). The lncRNA NBR2 was recently reported as an oncogene, whose function in CRC remains uncertain. The present study aimed to investigate the biological function of lncRNA NBR2 in the progression of CRC and its underlying molecular mechanisms. Ten pairs of clinical CRC and para-carcinoma tissues were collected to determine the expression levels of lncRNA NBR2 and miR-19a, and the polarization state of TAMs. Quantitative reverse transcriptase-polymerase chain reaction was used to evaluate the expression of miR-19a, and western blotting was used to determine the expression levels of tumor necrosis factor-α, human leukocyte antigen-DR, arginase-1, CD163, CD206, interleukin-4, AMP-activated protein kinase (AMPK), p-AMPK, hypoxia-inducible factor-1α (HIF-1α), protein kinase B (AKT), p-AKT, mechanistic target of rapamycin (mTOR), and p-mTOR in TAMs. The proliferative ability of HCT-116 cells was detected using the CCK8 assay, and the migratory ability of HCT-116 cells was evaluated using the Transwell assay. The interaction between lncRNA NBR2 and miR-19a was determined using the luciferase assay. The lncRNA NBR2 was downregulated and miR-19a was highly expressed in CRC cells, accompanied by a high M2 polarization. Downregulated miR-19a promoted M1 polarization, activated AMPK, suppressed HIF-1α and AKT/mTOR signaling pathways, and promoted antitumor properties in NBR2-overexpressed TAMs, which were all reversed by the introduction of the miR-19a mimic. LncRNA NBR2 was verified to target miR-19a in macrophages according to the results of the luciferase assay. Collectively, lncRNA NBR2 may suppress the progression of CRC by downregulating miR-19a to regulate M2 macrophage polarization.
Collapse
Affiliation(s)
- Xiaoting Yang
- School of Medicine, Quzhou College of Technology, Quzhou, Zhejiang, China
| | - Ye Luo
- School of Medicine, Quzhou College of Technology, Quzhou, Zhejiang, China
| | - Mengying Li
- School of Medicine, Quzhou College of Technology, Quzhou, Zhejiang, China
| | - Zhan Jin
- School of Medicine, Quzhou College of Technology, Quzhou, Zhejiang, China
| | - Gao Chen
- School of Medicine, Quzhou College of Technology, Quzhou, Zhejiang, China
| | - Chunchun Gan
- School of Medicine, Quzhou College of Technology, Quzhou, Zhejiang, China
| |
Collapse
|
12
|
Yao S, Han Y, Yang M, Jin K, Lan H. It's high-time to re-evaluate the value of induced-chemotherapy for reinforcing immunotherapy in colorectal cancer. Front Immunol 2023; 14:1241208. [PMID: 37920463 PMCID: PMC10619163 DOI: 10.3389/fimmu.2023.1241208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 10/09/2023] [Indexed: 11/04/2023] Open
Abstract
Immunotherapy has made significant advances in the treatment of colorectal cancer (CRC), revolutionizing the therapeutic landscape and highlighting the indispensable role of the tumor immune microenvironment. However, some CRCs have shown poor response to immunotherapy, prompting investigation into the underlying reasons. It has been discovered that certain chemotherapeutic agents possess immune-stimulatory properties, including the induction of immunogenic cell death (ICD), the generation and processing of non-mutated neoantigens (NM-neoAgs), and the B cell follicle-driven T cell response. Based on these findings, the concept of inducing chemotherapy has been introduced, and the combination of inducing chemotherapy and immunotherapy has become a standard treatment option for certain cancers. Clinical trials have confirmed the feasibility and safety of this approach in CRC, offering a promising method for improving the efficacy of immunotherapy. Nevertheless, there are still many challenges and difficulties ahead, and further research is required to optimize its use.
Collapse
Affiliation(s)
- Shiya Yao
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Yuejun Han
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Mengxiang Yang
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Huanrong Lan
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
13
|
Kiaie SH, Salehi-Shadkami H, Sanaei MJ, Azizi M, Shokrollahi Barough M, Nasr MS, Sheibani M. Nano-immunotherapy: overcoming delivery challenge of immune checkpoint therapy. J Nanobiotechnology 2023; 21:339. [PMID: 37735656 PMCID: PMC10512572 DOI: 10.1186/s12951-023-02083-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/26/2023] [Indexed: 09/23/2023] Open
Abstract
Immune checkpoint (ICP) molecules expressed on tumor cells can suppress immune responses against tumors. ICP therapy promotes anti-tumor immune responses by targeting inhibitory and stimulatory pathways of immune cells like T cells and dendritic cells (DC). The investigation into the combination therapies through novel immune checkpoint inhibitors (ICIs) has been limited due to immune-related adverse events (irAEs), low response rate, and lack of optimal strategy for combinatorial cancer immunotherapy (IMT). Nanoparticles (NPs) have emerged as powerful tools to promote multidisciplinary cooperation. The feasibility and efficacy of targeted delivery of ICIs using NPs overcome the primary barrier, improve therapeutic efficacy, and provide a rationale for more clinical investigations. Likewise, NPs can conjugate or encapsulate ICIs, including antibodies, RNAs, and small molecule inhibitors. Therefore, combining the drug delivery system (DDS) with ICP therapy could provide a profitable immunotherapeutic strategy for cancer treatment. This article reviews the significant NPs with controlled DDS using current data from clinical and pre-clinical trials on mono- and combination IMT to overcome ICP therapeutic limitations.
Collapse
Affiliation(s)
- Seyed Hossein Kiaie
- Department of Formulation Development, ReNAP Therapeutics, Tehran, Iran.
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Hossein Salehi-Shadkami
- Department of Formulation Development, ReNAP Therapeutics, Tehran, Iran
- Department of Medical Science, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Sanaei
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, 8815713471, Iran
| | - Marzieh Azizi
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | | | - Mohammad Sadegh Nasr
- Department of Computer Science and Engineering Multi-Interprofessional Center for Health Informatics (MICHI), The University of Texas at Arlington, Arlington, TX, USA
| | - Mohammad Sheibani
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Razi Drug Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Hexun Z, Miyake T, Maekawa T, Mori H, Yasukawa D, Ohno M, Nishida A, Andoh A, Tani M. High abundance of Lachnospiraceae in the human gut microbiome is related to high immunoscores in advanced colorectal cancer. Cancer Immunol Immunother 2023; 72:315-326. [PMID: 35869338 PMCID: PMC10991469 DOI: 10.1007/s00262-022-03256-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 07/09/2022] [Indexed: 01/26/2023]
Abstract
INTRODUCTION The tumor microenvironment (TME) in colorectal cancer (CRC) includes the gut microbiome, immune cells, angiogenic factors, and fibroblasts and plays a major role in cancer progression. The Immunoscore (IS) is based on tumor infiltration by immune cells that are known prognostic biomarkers for CRC. However, the interrelation between the IS, microbiome, and other TME factors in human CRC remains unclear. PATIENTS AND METHODS A cohort of 94 patients with CRC was examined at the Shiga University of Medical Science Hospital in Japan. The expression levels of CD3, CD8, CD31, and alpha-smooth muscle actin (α-SMA) in the primary tumor were evaluated by immunohistochemistry. The IS was calculated based on the results of the CD3 and CD8 staining assays. Microbiomes in patients with CRC were examined by amplicon sequencing. RESULTS The expression levels of α-SMA and tumor-infiltrating lymphocytes in patients with CRC were negatively correlated (P = 0.006). A high IS was associated with high abundance of Lachnospiraceae in the microbiomes of patients with CRC. CONCLUSION Lymphocyte infiltration into the primary tumor was marked by reduced density of cancer-associated fibroblasts and enrichment of the Lachnospiraceae family in the gut microbiome, which may influence CRC progression.
Collapse
Affiliation(s)
- Zhang Hexun
- Department of Surgery, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu City, Shiga, 520-2192, Japan
| | - Toru Miyake
- Department of Surgery, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu City, Shiga, 520-2192, Japan.
| | - Takeru Maekawa
- Department of Surgery, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu City, Shiga, 520-2192, Japan
| | - Haruki Mori
- Department of Surgery, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu City, Shiga, 520-2192, Japan
| | - Daiki Yasukawa
- Department of Surgery, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu City, Shiga, 520-2192, Japan
| | - Masashi Ohno
- Department of Gastroenterology and Hepatology, Nagahama City Hospital, Nagahama, Shiga, 5268580, Japan
- Department of Intestinal Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Atsushi Nishida
- Department of Intestinal Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Akira Andoh
- Department of Intestinal Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Masaji Tani
- Department of Surgery, Shiga University of Medical Science, Seta Tsukinowa-Cho, Otsu City, Shiga, 520-2192, Japan
| |
Collapse
|
15
|
Ghafelehbashi R, Farshbafnadi M, Aghdam NS, Amiri S, Salehi M, Razi S. Nanoimmunoengineering strategies in cancer diagnosis and therapy. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:78-90. [PMID: 36076122 DOI: 10.1007/s12094-022-02935-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/22/2022] [Indexed: 01/07/2023]
Abstract
Cancer immunotherapy strategies in combination with engineered nanosystems have yielded beneficial results in the treatment of cancer and their application is increasing day by day. The pivotal role of stimuli-responsive nanosystems and nanomedicine-based cancer immunotherapy, as a subsidiary discipline in the field of immunology, cannot be ignored. Today, rapid advances in nanomedicine are used as a platform for exploring new therapeutic applications and modern smart healthcare management strategies. The progress of nanomedicine in cancer treatment has confirmed the findings of immunotherapy in the medical research phase. This study concentrates on approaches connected to the efficacy of nanoimmunoengineering strategies for cancer immunotherapies and their applications. By assessing improved approaches, different aspects of the nanoimmunoengineering strategies for cancer therapies are discussed in this study.
Collapse
Affiliation(s)
- Robabehbeygom Ghafelehbashi
- Department of Materials and Textile Engineering, College of Engineering, Razi University, Kermanshah, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Melina Farshbafnadi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Shahin Amiri
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.,Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Mitra Salehi
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran. .,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran. .,School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Identification of immune subtypes and their prognosis and molecular implications in colorectal cancer. PLoS One 2022; 17:e0278114. [PMID: 36417424 PMCID: PMC9683557 DOI: 10.1371/journal.pone.0278114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 10/12/2022] [Indexed: 11/25/2022] Open
Abstract
Immune composition is commonly heterogeneous and varies among colorectal cancer (CRC) patients. A comprehensive immune classification may act as important characteristics to predict CRC prognosis. Thus, we aimed to identify novel immune specific subtypes to guide future therapies. Unsupervised clustering was used to classify CRC samples into different immune subtypes based on abundances of immune cell populations, during which TCGA and GSE17536 datasets were used as training and validation sets, respectively. The associations between the immune subtypes and patient prognosis were investigated. Further, we identified differentially expressed genes (DEGs) between immune high and low subtypes, followed by functional enrichment analyses of DEGs. The expression levels of 74 immunomodulators (IMs) across immune subtypes were analyzed. As a result, we clustered CRC samples into three distinct immune subtypes (immune high, moderate, and low). Patients with immune-high subtype showed the best prognosis, and patients with immune-low subtype had the worst survival in both TCGA and GSE17536 cohorts. A group of 2735 up-regulated DEGs were identified across immune high and low subtypes. The main DEGs were the members of complement components, chemokines, immunoglobulins, and immunosuppressive genes that are involved in immune modulation-related pathways (e.g., cytokine-cytokine receptor interaction) or GO terms (e.g., adaptive immune response and T cell activation). The expression levels of 63 IMs were significantly varied across immune subtypes. In conclusion, this study provides a conceptual framework and molecular characteristics of CRC immune subtypes, which may accurately predict prognosis and offer novel targets for personalized immunotherapy through modifying subtype-specific tumor immune microenvironment.
Collapse
|
17
|
Liu S, Zhang J, Zhang NN, Meng X, Liu K, Yang YG, Sun T, Sun L. Vibratome sectioning of tumors to evaluate the interactions between nanoparticles and the tumor microenvironment ex-vivo. Front Bioeng Biotechnol 2022; 10:1007151. [PMID: 36213072 PMCID: PMC9537459 DOI: 10.3389/fbioe.2022.1007151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 09/05/2022] [Indexed: 12/04/2022] Open
Abstract
Nanoparticles have been investigated as drug carriers and promising agents for cancer therapy. However, the tumor microenvironment (TME), which is formed by the tumor, is considered a barrier for nanocarriers to enter the internal tumor tissue. Therefore, the evaluation of the biological distribution of nanocarriers in TME can provide useful information on their role in tumor-targeted drug delivery. Although the tumor-bearing mouse model is commonly used to investigate the distribution of nanocarriers in the TME, there is currently a lack of a testing system to predict the distribution of nanocarriers in tumor tissues, especially in patients. This study revealed that the macrophages and dendritic cells (DCs) were more distributed in the peripheral part than the central part of the tumor, which might be an obstacle to the uniform distribution of nanoparticles in the tumor. In addition, the cellular uptake of gold nanoparticles (AuNR and AuNS) in macrophages and DCs cell lines (RAW264.7 and DC1.2) was markedly different from that in the TME. Hence, the study model of the interaction between nanoparticles and macrophages and DCs has an important impact on the accuracy of the results. The vibratome sections of tumor tissues preserved the spatial distribution of immune cells and tumor cells, and had very little effects on their morphologies and activities. More importantly, we found that the distribution of nanocarriers in vibratome sections was similar to that in tumors in vivo. In all, ex vivo analysis using vibratome sections of tumor tissues provides a more convenient and stable method for elucidating the influences of TME on the distribution of nanocarriers.
Collapse
Affiliation(s)
- Shuhan Liu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Juechao Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Ning-Ning Zhang
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, China
| | - Xiandi Meng
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
| | - Kun Liu
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- International Center of Future Science at Jilin University, Changchun, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, China
- International Center of Future Science at Jilin University, Changchun, China
| | - Liguang Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- *Correspondence: Liguang Sun,
| |
Collapse
|
18
|
Abdelgalil RM, Elmorshedy YM, Elkhodairy KA, Teleb M, Bekhit AA, Khattab SN, Elzoghby AO. Engineered nanomedicines for augmenting the efficacy of colorectal cancer immunotherapy. Nanomedicine (Lond) 2022; 17:1721-1745. [PMID: 36621872 DOI: 10.2217/nnm-2022-0214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most devastating diseases worldwide. Immunotherapeutic agents for CRC treatment have shown limited efficacy due to the immunosuppressive tumor microenvironment (TME). In this context, various types of nanoparticles (NPs) have been used to reverse the immunosuppressive TME, potentiate the effect of immunotherapeutic agents and reduce their systemic side effects. Many advantages could be offered by NPs, related to drug-loading efficiency, particle size and others that can potentially aid the delivery of immunotherapeutic agents. The recent research on how nano-based immunotherapy can remodel the immunosuppressive TME of CRC and hence boost the antitumor immune response, as well as the challenges that face clinical translation of NPs and future perspectives, are summarized in this review article.
Collapse
Affiliation(s)
- Riham M Abdelgalil
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, 21521, Alexandria, Egypt.,Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, 21521, Alexandria, Egypt
| | - Yomna M Elmorshedy
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, 21521, Alexandria, Egypt.,Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, 21521, Alexandria, Egypt
| | - Kadria A Elkhodairy
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, 21521, Alexandria, Egypt.,Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, 21521, Alexandria, Egypt
| | - Mohamed Teleb
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, 21521, Alexandria, Egypt.,Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, 21521, Alexandria, Egypt
| | - Adnan A Bekhit
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, 21521, Alexandria, Egypt.,Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, 21521, Alexandria, Egypt.,Pharmacy Program, Allied Health Department, College of Health & Sport Sciences, University of Bahrain, 32038, Riffa, Kingdom of Bahrain
| | - Sherine N Khattab
- Chemistry Department, Faculty of Science, Alexandria University, 21521, Alexandria, Egypt
| | - Ahmed O Elzoghby
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, 21521, Alexandria, Egypt.,Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, 21521, Alexandria, Egypt.,Division of Engineering in Medicine, Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, MA 02115, Boston, USA
| |
Collapse
|
19
|
Liu C, Liu D, Wang F, Xie J, Liu Y, Wang H, Rong J, Xie J, Wang J, Zeng R, Zhou F, Peng J, Xie Y. Identification of a glycolysis- and lactate-related gene signature for predicting prognosis, immune microenvironment, and drug candidates in colon adenocarcinoma. Front Cell Dev Biol 2022; 10:971992. [PMID: 36081904 PMCID: PMC9445192 DOI: 10.3389/fcell.2022.971992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 07/28/2022] [Indexed: 11/26/2022] Open
Abstract
Background: Colon adenocarcinoma (COAD), a malignant gastrointestinal tumor, has the characteristics of high mortality and poor prognosis. Even in the presence of oxygen, the Warburg effect, a major metabolic hallmark of almost all cancer cells, is characterized by increased glycolysis and lactate fermentation, which supports biosynthesis and provides energy to sustain tumor cell growth and proliferation. However, a thorough investigation into glycolysis- and lactate-related genes and their association with COAD prognosis, immune cell infiltration, and drug candidates is currently lacking. Methods: COAD patient data and glycolysis- and lactate-related genes were retrieved from The Cancer Genome Atlas (TCGA) and Gene Set Enrichment Analysis (GSEA) databases, respectively. After univariate Cox regression analysis, a nonnegative matrix factorization (NMF) algorithm was used to identify glycolysis- and lactate-related molecular subtypes. Least absolute shrinkage and selection operator (LASSO) Cox regression identified twelve glycolysis- and lactate-related genes (ADTRP, ALDOB, APOBEC1, ASCL2, CEACAM7, CLCA1, CTXN1, FLNA, NAT2, OLFM4, PTPRU, and SNCG) related to prognosis. The median risk score was employed to separate patients into high- and low-risk groups. The prognostic efficacy of the glycolysis- and lactate-related gene signature was assessed using Kaplan–Meier (KM) survival and receiver operating characteristic (ROC) curve analyses. The nomogram, calibration curves, decision curve analysis (DCA), and clinical impact curve (CIC) were employed to improve the clinical applicability of the prognostic signature. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed on differentially expressed genes (DEGs) from the high- and low-risk groups. Using CIBERSORT, ESTIMATE, and single-sample GSEA (ssGSEA) algorithms, the quantities and types of tumor-infiltrating immune cells were assessed. The tumor mutational burden (TMB) and cytolytic (CYT) activity scores were calculated between the high- and low-risk groups. Potential small-molecule agents were identified using the Connectivity Map (cMap) database and validated by molecular docking. To verify key core gene expression levels, quantitative real-time polymerase chain reaction (qRT–PCR) assays were conducted. Results: We identified four distinct molecular subtypes of COAD. Cluster 2 had the best prognosis, and clusters 1 and 3 had poor prognoses. High-risk COAD patients exhibited considerably poorer overall survival (OS) than low-risk COAD patients. The nomogram precisely predicted patient OS, with acceptable discrimination and excellent calibration. GO and KEGG pathway enrichment analyses of DEGs revealed enrichment mainly in the “glycosaminoglycan binding,” “extracellular matrix,” “pancreatic secretion,” and “focal adhesion” pathways. Patients in the low-risk group exhibited a larger infiltration of memory CD4+ T cells and dendritic cells and a better prognosis than those in the high-risk group. The chemotherapeutic agent sensitivity of patients categorized by risk score varied significantly. We predicted six potential small-molecule agents binding to the core target of the glycolysis- and lactate-related gene signature. ALDOB and APOBEC1 mRNA expression was increased in COAD tissues, whereas CLCA1 and OLFM4 mRNA expression was increased in normal tissues. Conclusion: In summary, we identified molecular subtypes of COAD and developed a glycolysis- and lactate-related gene signature with significant prognostic value, which benefits COAD patients by informing more precise and effective treatment decisions.
Collapse
Affiliation(s)
- Cong Liu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi, China
- Jiangxi Clinical Research Center for Gastroenterology, Nanchang, China
| | - Dingwei Liu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi, China
- Jiangxi Clinical Research Center for Gastroenterology, Nanchang, China
| | - Fangfei Wang
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi, China
- Jiangxi Clinical Research Center for Gastroenterology, Nanchang, China
| | - Jun Xie
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi, China
- Jiangxi Clinical Research Center for Gastroenterology, Nanchang, China
| | - Yang Liu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi, China
- Jiangxi Clinical Research Center for Gastroenterology, Nanchang, China
| | - Huan Wang
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi, China
- Jiangxi Clinical Research Center for Gastroenterology, Nanchang, China
| | - Jianfang Rong
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi, China
- Jiangxi Clinical Research Center for Gastroenterology, Nanchang, China
| | - Jinliang Xie
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi, China
- Jiangxi Clinical Research Center for Gastroenterology, Nanchang, China
| | - Jinyun Wang
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi, China
- Jiangxi Clinical Research Center for Gastroenterology, Nanchang, China
| | - Rong Zeng
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi, China
- Jiangxi Clinical Research Center for Gastroenterology, Nanchang, China
| | - Feng Zhou
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi, China
- Jiangxi Clinical Research Center for Gastroenterology, Nanchang, China
| | - Jianxiang Peng
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi, China
- Jiangxi Clinical Research Center for Gastroenterology, Nanchang, China
| | - Yong Xie
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Gastroenterology Institute of Jiangxi Province, Nanchang, Jiangxi, China
- Key Laboratory of Digestive Diseases of Jiangxi Province, Nanchang, Jiangxi, China
- Jiangxi Clinical Research Center for Gastroenterology, Nanchang, China
- *Correspondence: Yong Xie,
| |
Collapse
|
20
|
Zafari N, Khosravi F, Rezaee Z, Esfandyari S, Bahiraei M, Bahramy A, Ferns GA, Avan A. The role of the tumor microenvironment in colorectal cancer and the potential therapeutic approaches. J Clin Lab Anal 2022; 36:e24585. [PMID: 35808903 PMCID: PMC9396196 DOI: 10.1002/jcla.24585] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/07/2022] [Accepted: 06/23/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Colorectal cancer (CRC) with a high prevalence is recognized as the fourth most common cause of cancer-related death globally. Over the past decade, there has been growing interest in the network of tumor cells, stromal cells, immune cells, blood vessel cells, and fibroblasts that comprise the tumor microenvironment (TME) to identify new therapeutic interventions. METHODS Databases, such as Google Scholar, PubMed, and Scopus, were searched to provide an overview of the recent research progress related to targeting the TME as a novel therapeutic approach. RESULTS Tumor microenvironment as a result of the cross talk between these cells may result in either advantages or disadvantages in tumor development and metastasis, affecting the signals and responses from the surrounding cells. Whilst chemotherapy has led to an improvement in CRC patients' survival, the metastatic aspect of the disease remains difficult to avoid. CONCLUSIONS The present review emphasizes the structure and function of the TME, alterations in the TME, its role in the incidence and progression of CRC, the effects on tumor development and metastasis, and also the potential of its alterations as therapeutic targets. It should be noted that providing novel studies in this field of research might help us to achieve practical therapeutic strategies based on their interaction.
Collapse
Affiliation(s)
- Narges Zafari
- Department of Medical Genetics, School of MedicineTehran University of Medical SciencesTehranIran
| | - Fatemeh Khosravi
- Molecular Medicine Research Center, Hormozgan Health InstituteHormozgan University of Medical SciencesBandar AbbasIran
| | - Zahra Rezaee
- Department of Medical Genetics, Faculty of Medical SciencesTarbiat Modares UniversityTehranIran
| | - Sahar Esfandyari
- Department of Anatomy, School of MedicineTehran University of Medical SciencesTehranIran
| | - Mohamad Bahiraei
- Department of Radiology, Besat HospitalHamedan University of Medical SciencesHamedanIran
| | - Afshin Bahramy
- Department of Medical Genetics, Faculty of Medical SciencesTarbiat Modares UniversityTehranIran
| | - Gordon A. Ferns
- Brighton & Sussex Medical SchoolDivision of Medical EducationSussexUK
| | - Amir Avan
- Metabolic Syndrome Research CenterMashhad University of Medical SciencesMashhadIran
- Basic Medical Sciences InstituteMashhad University of Medical SciencesMashhadIran
- Medical Genetics Research CenterMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
21
|
Qiao C, Wang H, Guan Q, Wei M, Li Z. Ferroptosis-based nano delivery systems targeted therapy for colorectal cancer: Insights and future perspectives. Asian J Pharm Sci 2022; 17:613-629. [PMID: 36382305 PMCID: PMC9640473 DOI: 10.1016/j.ajps.2022.09.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/29/2022] [Accepted: 09/19/2022] [Indexed: 11/02/2022] Open
Abstract
There are limited options for patients who develop liver metastasis from colorectal cancer (CRC), the leading cause of cancer-related mortality worldwide. Emerging evidence has provided insights into iron deficiency and excess in CRC. Ferroptosis is an iron-dependent form of programmed cell death characterized by aberrant iron and lipid metabolism, which play crucial roles in tumorigenesis, tumor progression, and treatment options. A better understanding of the underlying molecular mechanism of ferroptosis has shed light on the current findings of ferroptosis-based nanodrug targeting strategies, such as driving ferroptosis in tumor cells and the tumor microenvironment, emerging combination therapy and against multidrug resistance. Furthermore, this review highlights the challenge and perspective of a ferroptosis-driven nanodrug delivery system for CRC-targeted therapy.
Collapse
Affiliation(s)
- Chu Qiao
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Haiying Wang
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Qiutong Guan
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Minjie Wei
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Zhenhua Li
- School of Pharmacy, China Medical University, Shenyang 110122, China
| |
Collapse
|
22
|
Chen F, Liu Q. Demystifying phytoconstituent-derived nanomedicines in their immunoregulatory and therapeutic roles in inflammatory diseases. Adv Drug Deliv Rev 2022; 186:114317. [PMID: 35533788 DOI: 10.1016/j.addr.2022.114317] [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: 03/31/2021] [Revised: 04/15/2022] [Accepted: 04/30/2022] [Indexed: 11/28/2022]
Abstract
In the past decades, phytoconstituents have appeared as critical mediators for immune regulations among various diseases, both in eukaryotes and prokaryotes. These bioactive molecules, showing a broad range of biological functions, would hold tremendous promise for developing new therapeutics. The discovery of phytoconstituents' capability of functionally regulating immune cells and associating cytokines, suppressing systemic inflammation, and remodeling immunity have rapidly promoted the idea of their employment as anti-inflammatory agents. In this review, we discuss various roles of phyto-derived medicines in the field of inflammatory diseases, including chronic inflammation, autoimmune diseases, and acute inflammatory disease such as COVID-19. Nevertheless, traditional phyto-derived medicines often concurred with their clinical administration limitations, such as their lack of cell specificity, inefficient cytoplasmic delivery, and rapid clearance by the immune system. As alternatives, phyto-derived nano-approaches may provide significant benefits. Both unmodified and engineered nanocarriers present the potential to serve as phytoconstituent delivery systems to improve therapeutic physio-chemical properties and pharmacokinetic profiles. Thus, the development of phytoconstituents' nano-delivery designs, their new and perspective approaches for therapeutical applications are elaborated herein.
Collapse
Affiliation(s)
- Fengqian Chen
- Translational Research Program, Department of Anesthesiology and Center for Shock Trauma Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Qi Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Cancer Research Building II, Suite 216, 1550 Orleans Street, Baltimore, MD 21231, United States.
| |
Collapse
|
23
|
Small Extracellular Vesicles: Key Forces Mediating the Development and Metastasis of Colorectal Cancer. Cells 2022; 11:cells11111780. [PMID: 35681475 PMCID: PMC9179504 DOI: 10.3390/cells11111780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 12/03/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide, and its incidence and mortality rates have been increasing annually in recent years. A variety of different small extracellular vesicles (sEVs) are important mediators of intercellular communication and have an important role in tumor metastasis and progression. The development and metastasis of CRC are closely linked to tumor-cell-derived sEVs, non-tumor-cell-derived sEVs, and intestinal-microbiota-derived sEVs. Numerous studies have shown that the tumor microenvironment (TME) is a key component in the regulation of CRC proliferation, development, and metastasis. These sEVs can create a TME conducive to CRC growth and metastasis by forming an immunosuppressive microenvironment, remodeling the extracellular matrix, and promoting tumor cell metabolism. Therefore, in this paper, we review the role of different types of sEVs in colorectal cancer development and metastasis. Furthermore, based on the properties of sEVs, we further discuss the use of sEVs as early biomarkers for colorectal cancer diagnosis and the potential for their use in the treatment of CRC.
Collapse
|
24
|
Mitarotonda R, Giorgi E, Eufrasio-da-Silva T, Dolatshahi-Pirouz A, Mishra YK, Khademhosseini A, Desimone MF, De Marzi M, Orive G. Immunotherapeutic nanoparticles: From autoimmune disease control to the development of vaccines. BIOMATERIALS ADVANCES 2022; 135:212726. [PMID: 35475005 PMCID: PMC9023085 DOI: 10.1016/j.bioadv.2022.212726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 02/09/2022] [Accepted: 02/18/2022] [Indexed: 11/01/2022]
Abstract
The development of nanoparticles (NPs) with potential therapeutic uses represents an area of vast interest in the scientific community during the last years. Recently, the pandemic caused by COVID-19 motivated a race for vaccines creation to overcome the crisis generated. This is a good demonstration that nanotechnology will most likely be the basis of future immunotherapy. Moreover, the number of publications based on nanosystems has significantly increased in recent years and it is expected that most of these developments can go on to experimentation in clinical stages soon. The therapeutic use of NPs to combat different diseases such as cancer, allergies or autoimmune diseases will depend on their characteristics, their targets, and the transported molecules. This review presents an in-depth analysis of recent advances that have been developed in order to obtain novel nanoparticulate based tools for the treatment of allergies, autoimmune diseases and for their use in vaccines. Moreover, it is highlighted that by providing targeted delivery an increase in the potential of vaccines to induce an immune response is expected in the future. Definitively, the here gathered analysis is a good demonstration that nanotechnology will be the basis of future immunotherapy.
Collapse
Affiliation(s)
- Romina Mitarotonda
- Laboratorio de Inmunología, Instituto de Ecología y Desarrollo Sustentable (INEDES) CONICET-UNLu, Departamento de Ciencias Básicas, Universidad Nacional de Luján, Ruta 5 y Avenida Constitución (6700) Lujan, Buenos Aires, Argentina
| | - Exequiel Giorgi
- Laboratorio de Inmunología, Instituto de Ecología y Desarrollo Sustentable (INEDES) CONICET-UNLu, Departamento de Ciencias Básicas, Universidad Nacional de Luján, Ruta 5 y Avenida Constitución (6700) Lujan, Buenos Aires, Argentina
| | - Tatiane Eufrasio-da-Silva
- Department of Health Technology, Technical University of Denmark (DTU), 2800 Kgs. Lyngby, Denmark; Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Department of Dentistry - Regenerative Biomaterials, Philips van Leydenlaan 25, 6525EX Nijmegen, the Netherlands
| | | | - Yogendra Kumar Mishra
- Mads Clausen Institute, NanoSYD, University of Southern Denmark, 6400 Sønderborg, Denmark
| | - Ali Khademhosseini
- Department of Bioengineering, Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA; Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA 90095, USA; Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA; Jonsson Comprehensive Cancer Center, Department of Radiology, University of California, Los Angeles, CA 90095, USA
| | - Martin F Desimone
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de la Química y Metabolismo del Fármaco (IQUIMEFA), Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina.
| | - Mauricio De Marzi
- Laboratorio de Inmunología, Instituto de Ecología y Desarrollo Sustentable (INEDES) CONICET-UNLu, Departamento de Ciencias Básicas, Universidad Nacional de Luján, Ruta 5 y Avenida Constitución (6700) Lujan, Buenos Aires, Argentina.
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain; Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore.
| |
Collapse
|
25
|
Barbălată CI, Porfire AS, Sesarman A, Rauca VF, Banciu M, Muntean D, Știufiuc R, Moldovan A, Moldovan C, Tomuță I. A Screening Study for the Development of Simvastatin-Doxorubicin Liposomes, a Co-Formulation with Future Perspectives in Colon Cancer Therapy. Pharmaceutics 2021; 13:pharmaceutics13101526. [PMID: 34683821 PMCID: PMC8537800 DOI: 10.3390/pharmaceutics13101526] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/08/2021] [Accepted: 09/15/2021] [Indexed: 01/25/2023] Open
Abstract
An increasing number of studies published so far have evidenced the benefits of Simvastatin (SIM) and Doxorubicin (DOX) co-treatment in colorectal cancer. In view of this, the current study aimed to investigate the pharmaceutical development of liposomes co-encapsulating SIM and DOX, by implementing the Quality by Design (QbD) concept, as a means to enhance the antiproliferative effect of the co-formulation on C26 murine colon cancer cells co-cultured with macrophages. It is known that the quality profile of liposomes is dependent on the critical quality attributes (CQAs) of liposomes (drug entrapped concentration, encapsulation efficiency, size, zeta potential, and drug release profile), which are, in turn, directly influenced by various formulation factors and processing parameters. By using the design of experiments, it was possible to outline the increased variability of CQAs in relation to formulation factors and identify by means of statistical analysis the material attributes that are critical (phospholipids, DOX and SIM concentration) for the quality of the co-formulation. The in vitro studies performed on a murine colon cancer cell line highlighted the importance of delivering the optimal drug ratio at the target site, since the balance antiproliferative vs. pro-proliferative effects can easily be shifted when the molar ratio between DOX and SIM changes.
Collapse
Affiliation(s)
- Cristina Ioana Barbălată
- Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Iuliu Hațieganu University of Medicine and Pharmacy, 41 Victor Babes Street, 400012 Cluj-Napoca, Romania; (C.I.B.); (D.M.); (I.T.)
| | - Alina Silvia Porfire
- Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Iuliu Hațieganu University of Medicine and Pharmacy, 41 Victor Babes Street, 400012 Cluj-Napoca, Romania; (C.I.B.); (D.M.); (I.T.)
- Correspondence:
| | - Alina Sesarman
- Department of Molecular Biology and Biotechnology, Centre for Systems Biology, Biodiversity and Bioresources (3B), Faculty of Biology and Geology, Babes-Bolyai University, 5-7 Clinicilor Street, 400006 Cluj-Napoca, Romania; (A.S.); (V.-F.R.); (M.B.)
- Molecular Biology Center, Institute for Interdisciplinary Research in Bio-Nano-Sciences of Babes-Bolyai University, 42 Treboniu Laurian Street, 400271 Cluj-Napoca, Romania
| | - Valentin-Florian Rauca
- Department of Molecular Biology and Biotechnology, Centre for Systems Biology, Biodiversity and Bioresources (3B), Faculty of Biology and Geology, Babes-Bolyai University, 5-7 Clinicilor Street, 400006 Cluj-Napoca, Romania; (A.S.); (V.-F.R.); (M.B.)
- Molecular Biology Center, Institute for Interdisciplinary Research in Bio-Nano-Sciences of Babes-Bolyai University, 42 Treboniu Laurian Street, 400271 Cluj-Napoca, Romania
| | - Manuela Banciu
- Department of Molecular Biology and Biotechnology, Centre for Systems Biology, Biodiversity and Bioresources (3B), Faculty of Biology and Geology, Babes-Bolyai University, 5-7 Clinicilor Street, 400006 Cluj-Napoca, Romania; (A.S.); (V.-F.R.); (M.B.)
- Molecular Biology Center, Institute for Interdisciplinary Research in Bio-Nano-Sciences of Babes-Bolyai University, 42 Treboniu Laurian Street, 400271 Cluj-Napoca, Romania
| | - Dana Muntean
- Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Iuliu Hațieganu University of Medicine and Pharmacy, 41 Victor Babes Street, 400012 Cluj-Napoca, Romania; (C.I.B.); (D.M.); (I.T.)
| | - Rareș Știufiuc
- MedFuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 4-6 Louis Pasteur Street, 400337 Cluj-Napoca, Romania; (R.Ș.); (A.M.); (C.M.)
| | - Alin Moldovan
- MedFuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 4-6 Louis Pasteur Street, 400337 Cluj-Napoca, Romania; (R.Ș.); (A.M.); (C.M.)
| | - Cristian Moldovan
- MedFuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 4-6 Louis Pasteur Street, 400337 Cluj-Napoca, Romania; (R.Ș.); (A.M.); (C.M.)
| | - Ioan Tomuță
- Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Iuliu Hațieganu University of Medicine and Pharmacy, 41 Victor Babes Street, 400012 Cluj-Napoca, Romania; (C.I.B.); (D.M.); (I.T.)
| |
Collapse
|
26
|
Li X, Zhu S, Yin P, Zhang S, Xu J, Zhang Q, Shi S, Zhang T. Combination immunotherapy of chlorogenic acid liposomes modified with sialic acid and PD-1 blockers effectively enhances the anti-tumor immune response and therapeutic effects. Drug Deliv 2021; 28:1849-1860. [PMID: 34515617 PMCID: PMC8439241 DOI: 10.1080/10717544.2021.1971797] [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: 11/29/2022] Open
Abstract
Melanoma is one of the most common malignant tumors. The anti-PD-1 antibody is used for the treatment of metastatic melanoma. Treatment success is only 35–40% and a range of immune-related adverse reactions can occur. Combination of anti-PD1 antibody therapy with other oncology therapies has been attempted. Herein, we assessed whether chlorogenic acid liposomes modified with sialic acid (CA-SAL) combined with anti-PD1 antibody treatment was efficacious as immunotherapy for melanoma. CA-SAL liposomes were prepared and characterized. In a mouse model of B16F10 tumor, mice were treated with an anti-PD1 antibody, CA-SAL, or combination of CA-SAL + anti-PD1 antibody, and compared with no treatment controls. The tumor inhibition rate, tumor-associated macrophages (TAMs) phenotype, T-cell activity, and safety were investigated. We observed a significant decrease in the proportion of M2-TAMs and CD4+Fop3+ T cells, while there was a significant increase in the proportion of M1-TAMs and CD8+ T cells, and in the activity of T cells, and thus in the tumor inhibition rate. No significant toxicity was observed in major organs. CA-SAL and anti-PD1 Ab combination therapy presented synergistic anti-tumor activity, which enhanced the efficacy of the PD-1 checkpoint blocker in a mouse model of melanoma. In summary, combination immunotherapy of CA-SAL and anti-PD1 Ab has broad prospects in improving the therapeutic effect of melanoma, and may provide a new strategy for clinical treatment.
Collapse
Affiliation(s)
- Xixi Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shunyao Zhu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ping Yin
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuangshuang Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Juewen Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qin Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Senlin Shi
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ting Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
27
|
Bashash D, Zandi Z, Kashani B, Pourbagheri-Sigaroodi A, Salari S, Ghaffari SH. Resistance to immunotherapy in human malignancies: Mechanisms, research progresses, challenges, and opportunities. J Cell Physiol 2021; 237:346-372. [PMID: 34498289 DOI: 10.1002/jcp.30575] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/19/2021] [Accepted: 08/24/2021] [Indexed: 12/31/2022]
Abstract
Despite remarkable advances in different types of cancer therapies, an effective therapeutic strategy is still a major and significant challenge. One of the most promising approaches in this regard is immunotherapy, which takes advantage of the patients' immune system; however, the many mechanisms that cancerous cells harbor to extend their survival make it impossible to gain perfect eradication of tumors. The response rate to cancer immunotherapies, especially checkpoint inhibitors and adoptive T cell therapy, substantially differs in various cancer types with the highest rates in advanced melanoma and non-small cell lung cancer. Indeed, the lack of response in many tumors indicates primary resistance that can originate from either tumor cells (intrinsic) or tumor microenvironment (extrinsic). On the other hand, some tumors show an initial response to immunotherapy followed by relapse in few months (acquired resistance). Understanding the underlying molecular mechanisms of immunotherapy resistance makes it possible to develop effective strategies to overcome this hurdle and boost therapy outcomes. In this review, we take a look at immunotherapy strategies and go through a number of primary and acquired resistance mechanisms. Also, we present various ongoing methods to overcoming resistance and introduce some promising fields to improve the outcome of immunotherapy in patients affected with cancer.
Collapse
Affiliation(s)
- Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Zandi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Kashani
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sina Salari
- Department of Medical Oncology, Hematology and Bone Marrow Transplantation, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed H Ghaffari
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Azwar S, Seow HF, Abdullah M, Faisal Jabar M, Mohtarrudin N. Recent Updates on Mechanisms of Resistance to 5-Fluorouracil and Reversal Strategies in Colon Cancer Treatment. BIOLOGY 2021; 10:854. [PMID: 34571731 PMCID: PMC8466833 DOI: 10.3390/biology10090854] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/04/2020] [Accepted: 08/11/2020] [Indexed: 02/06/2023]
Abstract
5-Fluorouracil (5-FU) plus leucovorin (LV) remain as the mainstay standard adjuvant chemotherapy treatment for early stage colon cancer, and the preferred first-line option for metastatic colon cancer patients in combination with oxaliplatin in FOLFOX, or irinotecan in FOLFIRI regimens. Despite treatment success to a certain extent, the incidence of chemotherapy failure attributed to chemotherapy resistance is still reported in many patients. This resistance, which can be defined by tumor tolerance against chemotherapy, either intrinsic or acquired, is primarily driven by the dysregulation of various components in distinct pathways. In recent years, it has been established that the incidence of 5-FU resistance, akin to multidrug resistance, can be attributed to the alterations in drug transport, evasion of apoptosis, changes in the cell cycle and DNA-damage repair machinery, regulation of autophagy, epithelial-to-mesenchymal transition, cancer stem cell involvement, tumor microenvironment interactions, miRNA dysregulations, epigenetic alterations, as well as redox imbalances. Certain resistance mechanisms that are 5-FU-specific have also been ascertained to include the upregulation of thymidylate synthase, dihydropyrimidine dehydrogenase, methylenetetrahydrofolate reductase, and the downregulation of thymidine phosphorylase. Indeed, the successful modulation of these mechanisms have been the game plan of numerous studies that had employed small molecule inhibitors, plant-based small molecules, and non-coding RNA regulators to effectively reverse 5-FU resistance in colon cancer cells. It is hoped that these studies would provide fundamental knowledge to further our understanding prior developing novel drugs in the near future that would synergistically work with 5-FU to potentiate its antitumor effects and improve the patient's overall survival.
Collapse
Affiliation(s)
- Shamin Azwar
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia; (S.A.); (H.F.S.); (M.A.)
| | - Heng Fong Seow
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia; (S.A.); (H.F.S.); (M.A.)
| | - Maha Abdullah
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia; (S.A.); (H.F.S.); (M.A.)
| | - Mohd Faisal Jabar
- Department of Surgery, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia;
| | - Norhafizah Mohtarrudin
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia; (S.A.); (H.F.S.); (M.A.)
| |
Collapse
|
29
|
Makaremi S, Asadzadeh Z, Hemmat N, Baghbanzadeh A, Sgambato A, Ghorbaninezhad F, Safarpour H, Argentiero A, Brunetti O, Bernardini R, Silvestris N, Baradaran B. Immune Checkpoint Inhibitors in Colorectal Cancer: Challenges and Future Prospects. Biomedicines 2021; 9:1075. [PMID: 34572263 PMCID: PMC8467932 DOI: 10.3390/biomedicines9091075] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy is a new pillar of cancer therapy that provides novel opportunities to treat solid tumors. In this context, the development of new drugs targeting immune checkpoints is considered a promising approach in colorectal cancer (CRC) treatment because it can be induce specific and durable anti-cancer effects. Despite many advances in the immunotherapy of CRC, there are still limitations and obstacles to successful treatment. The immunosuppressive function of the tumor microenvironment (TME) is one of the causes of poor response to treatment in CRC patients. For this reason, checkpoint-blocking antibodies have shown promising outcomes in CRC patients by blocking inhibitory immune checkpoints and enhancing immune responses against tumors. This review summarizes recent advances in immune checkpoint inhibitors (ICIs), such as CTLA-4, PD-1, PD-L1, LAG-3, and TIM-3 in CRC, and it discusses various therapeutic strategies with ICIs, including the double blockade of ICIs, combination therapy of ICIs with other immunotherapies, and conventional treatments. This review also delineates a new hopeful path in the combination of anti-PD-1/anti-PD-L1 with other ICIs such as anti-CTLA-4, anti-LAG-3, and anti-TIM-3 for CRC treatment.
Collapse
Affiliation(s)
- Shima Makaremi
- Department of Immunology & Microbiology, School of Medicine, Arak University of Medical Sciences, Arak 3848176941, Iran;
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (Z.A.); (N.H.); (A.B.); (F.G.)
| | - Nima Hemmat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (Z.A.); (N.H.); (A.B.); (F.G.)
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (Z.A.); (N.H.); (A.B.); (F.G.)
| | - Alessandro Sgambato
- Istituto di Ricovero e Cura a Carattere Scientifico Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 5972362 Rome, Italy;
- Area of Pathology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 5972362 Rome, Italy
| | - Farid Ghorbaninezhad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (Z.A.); (N.H.); (A.B.); (F.G.)
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
| | - Hossein Safarpour
- Cellular & Molecular Research Center, Birjand University of Medical Sciences, Birjand 9717853577, Iran;
| | - Antonella Argentiero
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; (A.A.); (O.B.)
| | - Oronzo Brunetti
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; (A.A.); (O.B.)
| | - Renato Bernardini
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95121 Catania, Italy;
| | - Nicola Silvestris
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; (A.A.); (O.B.)
- Department of Biomedical Sciences and Human Oncology (DIMO), University of Bari, 70124 Bari, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran; (Z.A.); (N.H.); (A.B.); (F.G.)
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz 5166/15731, Iran
| |
Collapse
|
30
|
Rizzo G, Bertotti A, Leto SM, Vetrano S. Patient-derived tumor models: a more suitable tool for pre-clinical studies in colorectal cancer. J Exp Clin Cancer Res 2021; 40:178. [PMID: 34074330 PMCID: PMC8168319 DOI: 10.1186/s13046-021-01970-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/02/2021] [Indexed: 12/15/2022] Open
Abstract
Colorectal cancer (CRC), despite the advances in screening and surveillance, remains the second most common cause of cancer death worldwide. The biological inadequacy of pre-clinical models to fully recapitulate the multifactorial etiology and the complexity of tumor microenvironment and human CRC's genetic heterogeneity has limited cancer treatment development. This has led to the development of Patient-derived models able to phenocopy as much as possible the original inter- and intra-tumor heterogeneity of CRC, reflecting the tumor microenvironment's cellular interactions. Implantation of patient tissue into immunodeficient mice hosts and the culture of tumor organoids have allowed advances in cancer biology and metastasis. This review highlights the advantages and limits of Patient-derived models as innovative and valuable pre-clinical tools to study progression and metastasis of CRC, develop novel therapeutic strategies by creating a drug screening platform, and predict the efficacy of clinical response to therapy.
Collapse
Affiliation(s)
- Giulia Rizzo
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, 20090, Milan, Italy
| | - Andrea Bertotti
- Laboratory of Translational Cancer Medicine, Candiolo Cancer Institute - FPO IRCCs, Candiolo, 10060, Torino, Italy
- Department of Oncology, University of Torino School of Medicine, Candiolo, 10060, Torino, Italy
| | - Simonetta Maria Leto
- Laboratory of Translational Cancer Medicine, Candiolo Cancer Institute - FPO IRCCs, Candiolo, 10060, Torino, Italy
| | - Stefania Vetrano
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, 20090, Milan, Italy.
- IBD Center, Department of Gastroenterology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy.
| |
Collapse
|
31
|
Torreggiani E, Bononi I, Pietrobon S, Mazzoni E, Guerra G, Feo C, Martini F, Tognon M. Colorectal Carcinoma Affected Patients Are Significantly Poor Responders Against the Oncogenic JC Polyomavirus. Front Immunol 2021; 12:632129. [PMID: 34113338 PMCID: PMC8185217 DOI: 10.3389/fimmu.2021.632129] [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: 11/22/2020] [Accepted: 05/07/2021] [Indexed: 12/24/2022] Open
Abstract
Background Many investigations reported the association between human tumors and JCPyV, a polyomavirus with oncogenic potential. The association has been supported by studies that found JCPyV footprints in CRC and gliomas of different types. Indeed, JCPyV footprints including its nucleic acids and Tag oncoprotein have been revealed in CRC tissues. Methods Herein, sera from colorectal carcinoma (CRC) affected patients and healthy individuals (HS), employed as control, were analysed for immunoglobulin G (IgG) antibodies against specific JCPyV viral capsid protein 1 (VP1) antigens. The investigation was carried out employing an innovative immunological assay. Indeed, an indirect enzyme-linked immunosorbent assay (ELISA) with JCPyV VP1 mimotopes was used. JCPyV VP1 mimotopes consisted of synthetic peptides mimicking VP1 epitopes. Results Sera from CRC affected patients, evaluated using indirect ELISAs with synthetic mimotopes, showed a significant lower prevalence of IgG antibodies against JCPyV VP1 mimotopes (26%) compared to HS (51%), p<0.005. These data were confirmed by another method, the hemagglutination inhibition (HAI) assay. Altogether these results, i.e. the prevalence of serum IgG antibodies against JCPyV VP1 mimotopes from patients with CRC is approximately 50% lower than in HS, are of interest. Discussion Our data suggest that patients with CRC are significantly poor responders against JCPyV VP1 antigens. It is possible that CRC patients are affected by a specific immunological deregulation. This immunological dysfunction, revelled in CRC patients, may account for their predisposition to the colorectal carcinoma onset.
Collapse
Affiliation(s)
- Elena Torreggiani
- Department of Medical Sciences, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Ilaria Bononi
- Department of Translational Medicine, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Silvia Pietrobon
- Department of Medical Sciences, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Elisa Mazzoni
- Department of Medical Sciences, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Giovanni Guerra
- Clinical Laboratory Analysis, University-Hospital of Ferrara, Ferrara, Italy
| | - Carlo Feo
- Department of Medical Sciences, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Fernanda Martini
- Department of Medical Sciences, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Mauro Tognon
- Department of Medical Sciences, School of Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
32
|
Silveira MJ, Castro F, Oliveira MJ, Sarmento B. Immunomodulatory nanomedicine for colorectal cancer treatment: a landscape to be explored? Biomater Sci 2021; 9:3228-3243. [PMID: 33949441 DOI: 10.1039/d1bm00137j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Colorectal cancer (CRC) is one of the deadliest cancers in the world mainly due to metastasis events. Despite improvements, the available treatment modalities for metastatic cases are limited, being generally associated with poor prognosis. As is well known, the immunosuppressive tumor microenvironment (TME) plays a key role in tumorigenesis, promoting cancer cell immune escape and disease progression. In addition, accumulating evidence indicates that the immunosuppressive microenvironment is a critical barrier for antitumor immunity in CRC, being extremely important to modulate the immune microenvironment to inhibit the tumor-promoting immune response. Therefore, new and effective cancer immunotherapeutic approaches demand a better control over the TME to reverse these immunosuppressive conditions. According to the features of different nanomedicines, nanoparticles can constitute a promising strategy, using different materials with the inherent ability to modulate TME and also with the potential to target immunosuppressive cells, to deliver antigens or immunomodulatory agents to eliminate this tumor. In this review, we summarize the importance of the TME in the progression and treatment response of CRC, exploring the potential of the nanotechnology for the development of immunomodulatory therapeutic strategies.
Collapse
Affiliation(s)
- Maria José Silveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal. and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal and ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Flávia Castro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal. and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Maria José Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal. and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal and FMUP - Departamento de Anatomia Patológica, Faculdade de Medicina, Universidade do Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal. and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal and CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| |
Collapse
|
33
|
Hanus M, Parada-Venegas D, Landskron G, Wielandt AM, Hurtado C, Alvarez K, Hermoso MA, López-Köstner F, De la Fuente M. Immune System, Microbiota, and Microbial Metabolites: The Unresolved Triad in Colorectal Cancer Microenvironment. Front Immunol 2021; 12:612826. [PMID: 33841394 PMCID: PMC8033001 DOI: 10.3389/fimmu.2021.612826] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/02/2021] [Indexed: 12/16/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide. As with other cancers, CRC is a multifactorial disease due to the combined effect of genetic and environmental factors. Most cases are sporadic, but a small proportion is hereditary, estimated at around 5-10%. In both, the tumor interacts with heterogeneous cell populations, such as endothelial, stromal, and immune cells, secreting different signals (cytokines, chemokines or growth factors) to generate a favorable tumor microenvironment for cancer cell invasion and metastasis. There is ample evidence that inflammatory processes have a role in carcinogenesis and tumor progression in CCR. Different profiles of cell activation of the tumor microenvironment can promote pro or anti-tumor pathways; hence they are studied as a key target for the control of cancer progression. Additionally, the intestinal mucosa is in close contact with a microorganism community, including bacteria, bacteriophages, viruses, archaea, and fungi composing the gut microbiota. Aberrant composition of this microbiota, together with alteration in the diet-derived microbial metabolites content (such as butyrate and polyamines) and environmental compounds has been related to CRC. Some bacteria, such as pks+ Escherichia coli or Fusobacterium nucleatum, are involved in colorectal carcinogenesis through different pathomechanisms including the induction of genetic mutations in epithelial cells and modulation of tumor microenvironment. Epithelial and immune cells from intestinal mucosa have Pattern-recognition receptors and G-protein coupled receptors (receptor of butyrate), suggesting that their activation can be regulated by intestinal microbiota and metabolites. In this review, we discuss how dynamics in the gut microbiota, their metabolites, and tumor microenvironment interplays in sporadic and hereditary CRC, modulating tumor progression.
Collapse
Affiliation(s)
- Michelle Hanus
- Laboratory of Innate Immunity, Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | - Daniela Parada-Venegas
- Laboratory of Innate Immunity, Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | - Glauben Landskron
- Laboratory of Innate Immunity, Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | | | - Claudia Hurtado
- Research Core, Academic Department, Clínica Las Condes, Santiago, Chile
| | - Karin Alvarez
- Cancer Center, Clínica Universidad de los Andes, Santiago, Chile
| | - Marcela A. Hermoso
- Laboratory of Innate Immunity, Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | | | | |
Collapse
|
34
|
Jakubowska K, Koda M, Grudzińska M, Lomperta K, Famulski W. Tumor-infiltrating lymphocytes in tissue material combined with systemic lymphocyte inflammation in patients with colorectal cancer. Mol Clin Oncol 2021; 14:97. [PMID: 33767866 DOI: 10.3892/mco.2021.2259] [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: 06/22/2020] [Accepted: 02/11/2021] [Indexed: 11/06/2022] Open
Abstract
The efficacy of cancer immunotherapy has been actively explored in the treatment of various malignant neoplasms of the gastrointestinal tract. In light of recent reports, the present study aimed to investigate the combination of the absolute lymphocyte count (ALC), percentage of tumor-infiltrating lymphocyte (TILs) and tumor progression status in patients with colorectal cancer (CRC) who underwent surgery. The medical records of 160 patients diagnosed with CRC were retrospectively reviewed. TILs were determined as a percentage of mononuclear inflammatory cells in the total intratumoral or stromal area as determined in five high power fields (magnification, x200-400), at the invasive front and in the centre of the tumour. Blood samples were obtained within 3 days prior to and 7 days following the surgical treatment. The assessment of the TIL percentage was performed in the tissue at the invasive front and in the centre of the primary tumour mass in combination with the determination of ALC in whole blood samples. The samples were obtained prior to and after surgery from patients with CRC, and the tumour progression status was also assessed (TILs/ALC/tumour progression status). A significant association was observed between the percentage of TILs in the main mass of tumour and the tumour size (P=0.031), the pT stage (P=0.049) and the incidence of necrosis (P=0.037) following surgery. The histological type was associated with the evaluated combined parameters prior to surgery (P=0.046). Lymph node pouch invasion was associated with TILs at the invasive front of tumour and with ALC prior to and after surgery (P=0.006 and P=0.037). Furthermore, the data indicated that the percentage of TILs located on the invasive front and centre of the tumour, and the ALC prior to and following surgery correlated with the treatment status (P=0.032, P=0.018, P≤0.001 and P≤0.001). A significant association was noted between eight features and evaluated combined parameters following surgery. These included the tumour size (P=0.021), TNM stage (P<0.001), tumour deposits (P=0.001), incidence of necrosis (P=0.042) and lymph node metastasis (P<0.001). Furthermore, the degree of invasion of venous (P<0.001), lymphatic (P<0.001) and perineural (P<0.001) sites was also significantly associated with TILs, ALC obtained after surgical treatment and tumor progression status. The data demonstrated that local and systemic chronic inflammation was associated with tumour progression in patients with CRC.
Collapse
Affiliation(s)
- Katarzyna Jakubowska
- Department of Pathomorphology, Comprehensive Cancer Center, 15-027 Bialystok, Poland
| | - Mariusz Koda
- Department of General Pathomorphology, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Małgorzata Grudzińska
- Department of General Pathomorphology, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Karolina Lomperta
- Department of Pathomorphology, Comprehensive Cancer Center, 15-027 Bialystok, Poland
| | - Waldemar Famulski
- Department of Medical Pathomorphology, Medical University of Bialystok, 15-269 Bialystok, Poland
| |
Collapse
|
35
|
Lu G, Qiu Y, Su X. Targeting CXCL12-CXCR4 Signaling Enhances Immune Checkpoint Blockade Therapy Against Triple Negative Breast Cancer. Eur J Pharm Sci 2021; 157:105606. [PMID: 33131745 DOI: 10.1016/j.ejps.2020.105606] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 06/21/2020] [Accepted: 10/16/2020] [Indexed: 12/18/2022]
Abstract
Insufficient T cell infiltration in triple-negative breast cancer (TNBC) has limited its response rate to immune checkpoint blockade (ICB) therapies and motivated the development of immunostimulatory approaches to enhance the ICB therapy. CXCR4 is a chemokine receptor highly upregulated both on cell surface and cytoplasm in tumor tissues. Activating CXCR4 has been associated with increased immunosuppression in the tumor microenvironment. Here, we developed a CXCR4-targeted liposomal formulation (Liposomal-AMD3100) to enhance therapeutic efficacy of AMD3100, a CXCR4 antagonist. Particularly, AMD3100 is not only encapsulated into the liposome but coated on the surface of the formulation to serve as a targeting moiety and a dual blocker capable of inhibiting CXCR4 activation extracellularly and intracellularly. The Liposomal-AMD3100 remodeled both immune and stromal microenvironment more efficiently compared with free AMD3100, indicating better pharmacodynamic profile of AMD3100 achieved by liposomal formulation. The combination of anti-PD-L1 with Liposomal-AMD3100 formulation exhibited an increased antitumor effect and prolonged survival time compared with monotherapies in a murine TNBC model (4T1). This work proves that immune activation via liposomal delivery of CXCR4 inhibitors has a great potential to expand ICB therapies to originally ICB-insensitive cancer types.
Collapse
Affiliation(s)
- Guowen Lu
- Department of Thyroid and breast mininally invasive surgery, Ningbo Yinzhou People's Hospital, No.251 Baizhang East Road, 315000 Ningbo, Zhejiang, P.R. China.
| | - Yier Qiu
- Department of Thyroid and breast mininally invasive surgery, Ningbo Yinzhou People's Hospital, No.251 Baizhang East Road, 315000 Ningbo, Zhejiang, P.R. China
| | - Xiaobao Su
- Department of Thyroid and breast mininally invasive surgery, Ningbo Yinzhou People's Hospital, No.251 Baizhang East Road, 315000 Ningbo, Zhejiang, P.R. China
| |
Collapse
|
36
|
Shahnazari M, Samadi P, Pourjafar M, Jalali A. Therapeutic vaccines for colorectal cancer: The progress and future prospect. Int Immunopharmacol 2020; 88:106944. [PMID: 33182032 DOI: 10.1016/j.intimp.2020.106944] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/21/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023]
Abstract
Cancer vaccines are usually derived from the patient's tumor cells or the antigens found on their surface, which may help the immune system to identify and kill these malignant cells. Current focus of many researches is designing vaccines with the hope of triggering the immune system to attack cancer cells in a more effective, reliable and safe manner. Although colorectal cancer (CRC) is recognized as the third leading cause of death by cancer, but significant advances in therapy strategies have been made in recent years, including cancer vaccine. In this review, we present various vaccine platforms that have been used in the border battle against CRC, some of which have been approved for clinical use and some are in late-stage clinical trials. Until September 2020 there is approximately 1940 clinical trials of cancer vaccines on patients with different cancer types, and also many more trials are in the planning stages, which makes it the most important period of therapeutic cancer vaccines studies in the history of the immunotherapy. In cancer vaccines clinical trials, there are several considerations that must be taken into account including engineering of antigen-presenting cells, potential toxicity of antigenic areas, pharmacokinetics and pharmacodynamics of vaccines, and monitoring of the patients' immune response. Therefore, the need to overcome immunosuppression mechanisms/immune tolerance is a critical step for the success of introducing therapeutic vaccines into the widely used drugs on market. In this way, better understanding of neoantigens, tumor immune surveillance escape mechanisms and host-tumor interactions are required to develop more effective and safe cancer vaccines.
Collapse
Affiliation(s)
- Mina Shahnazari
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Pouria Samadi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Mona Pourjafar
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Akram Jalali
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
37
|
Xiong Y, Song W, Shen L, Wang Y, Zhang J, Hu M, Liu Y, Li J, Musetti S, Liu R, Huang L. Oral Metformin and Polymetformin Reprogram Immunosuppressive Microenvironment and Boost Immune Checkpoint Inhibitor Therapy in Colorectal Cancer. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000168] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Yang Xiong
- UNC Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27559 USA
- College of Pharmaceutical Science Zhejiang Chinese Medical University Hangzhou Zhejiang 310053 China
| | - Wantong Song
- UNC Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27559 USA
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun Jilin 130022 China
| | - Limei Shen
- UNC Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27559 USA
| | - Ying Wang
- UNC Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27559 USA
| | - Jing Zhang
- UNC Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27559 USA
| | - Mengying Hu
- UNC Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27559 USA
| | - Yun Liu
- UNC Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27559 USA
| | - Jingjing Li
- UNC Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27559 USA
| | - Sara Musetti
- UNC Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27559 USA
| | - Rihe Liu
- UNC Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27559 USA
| | - Leaf Huang
- UNC Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27559 USA
| |
Collapse
|
38
|
Fu X, Luo H, Zheng Y, Wang S, Zhong Z, Wang Y, Yang Y. CTLA-4 immunotherapy exposes differences in immune response along with different tumor progression in colorectal cancer. Aging (Albany NY) 2020; 12:15656-15669. [PMID: 32805718 PMCID: PMC7467381 DOI: 10.18632/aging.103765] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/07/2020] [Indexed: 12/11/2022]
Abstract
Tumor growth is accompanied by a changing tumor microenvironment and mutations that increase the resistance to therapy. Here, we used syngeneic models to evaluate the drug response of tumors of the same type of different sizes. We used the in vivo efficacy and Ki-67 immunohistochemistry (IHC) assay to assess the difference in responses in response to treatment with the same concentration of anti-CTLA-4. Flow cytometry analysis revealed changes in the immune subpopulations changes the spleen, peripheral blood, lymph node, and tumor tissue across different tumor growth phases. For example, naive CD4+T, CD4+TCM, CD8+TEM, T, B, Treg, CD8+TCM exhibited different percentages depending on the specific immune organ. To further expose the changes in the immune microenvironment, the level of expression of PD-1 and CTLA-4 showed statistically significant difference in related subsets for each four immune tissues in different tumor sizes. In addition, the ratios of CD4 + Teff/ CD4 + Treg and CD8 + T/Treg in corresponding immune tissue were also associated with statistically significant differences alongside tumor growth in different animal models. These results reveal the ongoing changes in the immune microenvironment during tumor progression and anti-CTLA-4 antibody immunotherapy effect depends on the expression level of immune factors.
Collapse
Affiliation(s)
- Xiaocong Fu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
- Equal contribution
| | - Hua Luo
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
- Equal contribution
| | - Yuhui Zheng
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Shengpeng Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zhangfeng Zhong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yitao Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yeguo Yang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| |
Collapse
|
39
|
Lei S, Zhang X, Men K, Gao Y, Yang X, Wu S, Duan X, Wei Y, Tong R. Efficient Colorectal Cancer Gene Therapy with IL-15 mRNA Nanoformulation. Mol Pharm 2020; 17:3378-3391. [PMID: 32787272 DOI: 10.1021/acs.molpharmaceut.0c00451] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Immunogene therapy is a novel method for the treatment of colorectal cancer. Cytokine IL-15 has exhibited therapeutic anticancer potential due to its immune-stimulation property. However, conventional IL-15-based cancer gene therapy studies have been performed using the plasmid DNA form, which has potential shortcomings including weak delivery efficiency and backbone effect. In this study, an IL-15 immunogene therapy study for colon cancer using in vitro transcript mRNA is described. A protamine/liposome system (CLPP) is developed to provide efficient condensation and delivery capacity for in vivo mRNA transportation. They demonstrated that the prepared CLPP system could deliver the IL-15-encoding mRNA into C26 cells with high efficacy. The secretory expressed IL-15 cytokine by the C26 cells successfully produced lymphocyte stimulation and triggered anticancer cytotoxicity upon cancer cells in vitro. Local or systemic administration of the CLPP/mIL-15 complex exhibited obvious inhibition effects on multiple C26 murine colon cancer models with inhibition rates of up to 70% in the C26 abdominal cavity metastasis tumor model, 55% in the subcutaneous model, and 69% in the pulmonary metastasis model, demonstrating high efficacy and safety. These results successfully demonstrated the high therapeutic potential of the CLPP/mIL-15 complex for colorectal cancer immunogene therapy.
Collapse
Affiliation(s)
- Sibei Lei
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, People's Republic of China
| | - Xueyan Zhang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, People's Republic of China
| | - Ke Men
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, People's Republic of China
| | - Yan Gao
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, People's Republic of China
| | - Xijing Yang
- Animal Experiment Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Sisi Wu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, People's Republic of China
| | - Xingmei Duan
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, People's Republic of China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, People's Republic of China
| | - Rongsheng Tong
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, People's Republic of China
| |
Collapse
|
40
|
Zhao X, Weng W, Long Y, Pan W, Li Z, Sun F. LINC00665/miR-9-5p/ATF1 is a novel axis involved in the progression of colorectal cancer. Hum Cell 2020; 33:1142-1154. [PMID: 32776307 DOI: 10.1007/s13577-020-00393-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/17/2020] [Indexed: 02/08/2023]
Abstract
Long noncoding RNAs (lncRNAs) are abnormally expressed in many malignant tumors and involved in regulating the malignant phenotypes of cancer cells. However, the role of LINC00665 in colorectal cancer (CRC) and its regulatory mechanism remain unclear. In this study, real-time polymerase chain reaction (RT-PCR) was used to detect the expressions of LINC00665, miR-9-5p and activating transcription factor 1 (ATF1) mRNA in CRC tissues. The expression of ATF1 in CRC tissues was also detected by immunohistochemistry and Western blot. CCK-8 and colony formation assays were employed to detect cell proliferation. Cell cycle and apoptosis were detected by flow cytometry analysis. Scratch healing assay and Transwell test were exploited to detect cell migration and invasion. The targeting relationships between LINC00665 and miR-9-5p, and miR-9-5p and ATF1 were validated by dual luciferase reporter assay. We found that LINC00665 was significantly overexpressed in CRC tissues, and it was also negatively correlated with the expression of miR-9-5p and positively associated with the expression of ATF1. Besides, LINC00665 promoted the proliferation, migration and invasion of CRC cells, and inhibited cell apoptosis by sponging miR-9-5p. ATF1 was proved to be the downstream target of miR-9-5p and was indirectly regulated by LINC00665. Collectively, it is concluded that LINC00665 contributes to the progression of CRC by regulating miR-9-5p/ATF1 axis.
Collapse
Affiliation(s)
- Xuhong Zhao
- Department of Clinical Laboratory, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090, People's Republic of China
| | - Wenhao Weng
- Department of Clinical Laboratory, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090, People's Republic of China
| | - Yin Long
- Department of Clinical Laboratory, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090, People's Republic of China
| | - Weijie Pan
- Department of Clinical Laboratory, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090, People's Republic of China
| | - Zhi Li
- Department of Clinical Laboratory, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090, People's Republic of China.
| | - Fenyong Sun
- Department of Clinical Laboratory, Shanghai 10th People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai, 200072, People's Republic of China.
| |
Collapse
|
41
|
Musa M, Ali A. Cancer-associated fibroblasts of colorectal cancer and their markers: updates, challenges and translational outlook. Future Oncol 2020; 16:2329-2344. [PMID: 32687721 DOI: 10.2217/fon-2020-0384] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Accumulation of cancer-associated fibroblasts (CAFs) in the tumor microenvironment is associated with poor prognosis and recurrence of colorectal cancer (CRC). Despite their prominent roles in colorectal carcinogenesis, there is a lack of robust and specific markers to classify the heterogeneous and highly complex CAF populations. This has resulted in confusing and misleading definitions of CAFs in cancer niche. Advancements in molecular biology approaches have open doors to reliable CAF marker detection methods in various solid tumors. These discoveries would contribute to more efficient screening, monitoring and targeted therapy of CRC thus potentially will reduce cancer morbidity and mortality rates. This review highlights current scenarios, dilemma, translational potentials of CAF biomarker and future therapeutic applications involving CAF marker identification in CRC.
Collapse
Affiliation(s)
- Marahaini Musa
- Human Genome Centre, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Adli Ali
- Department of Paediatrics, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Bandar Tun Razak, Wilayah Persekutuan, 56000 Kuala Lumpur, Malaysia.,Department of Paediatrics, Oxford University, Level 2, Children's Hospital, John Radcliffe Hospital, Headington, Oxford OX3 9DU, UK
| |
Collapse
|
42
|
Yu LG, Chen SQ, Mao GL. Relationship between EZH2 and VEGF expression and prognosis in colorectal cancer. Shijie Huaren Xiaohua Zazhi 2020; 28:155-166. [DOI: 10.11569/wcjd.v28.i5.155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a common malignant tumor of the digestive system. However, its exact pathogenesis and independent prognostic factors are still unclear. In this study, we analyzed the expression of enhancer of zeste homolog 2 (EZH2) and vascular endothelial growth factor (VEGF) genes in CRC by using bioinformatics method and their relationship with prognosis. The expression of EZHE2 and VEGF proteins and their relationship with the patients' clinical characteristics were detected by immunohistochemistry.
AIM To investigate the expression and mutation status of EZH2 and VEGF in CRC and their relationship with clinicopathological features and prognosis.
METHODS The expression of EZH2 and VEGF genes in CRC and tumor adjacent tissues was compared in the TCGA database, and the mutations of EZH2 and VEGF genes were analyzed. The expression network of EZH2 and VEGF genes was established with the STRING database and the key genes in the network were screened. According to the expression of EZH2 and VEGF in tumor tissues, the patients were divided into high and low expression groups. Cox regression model and log-rank test were used to compare the difference of total survival and disease-free survival between the high and low expression groups. In addition, 80 patients with CRC who underwent surgery were selected, and their cancer tissues and adjacent tissues were collected. The expression of EZH2 and VEGF proteins in the above tissues was detected by immunohistochemistry.
RESULTS Analysis based on the TCGA database showed that the expression levels of EZH2 and VEGF genes in CRC tissues were significantly higher than those in matched normal intestinal epithelial tissues (P < 0.05), but they had no significant correlation with clinical stage of CRC (P > 0.05).The mutation rates of EZH2 and VEGF genes in human CRC were 1.5% and 1.9%, respectively, and there were significant differences in the expression levels of EZH2 and VEGF mRNA in tissues with different EZH2 and VEGF mutation statuses. There are 22 proteins in the network. The average interaction index of each protein is 10.5, the regional aggregation index is 0.8, and the protein enrichment was obvious (P < 0.01). EZH2, DNMT1, HDAC2, YY1, and SUZ12 were screened as the key genes in the network using Cytohubb software. There was no correlation between the expression of EZH2 and VEGF and the total survival time of patients (P > 0.05), but the disease-free survival time of patients with high expression of VEGF was significantly lower than that of patients with low expression of VEGF (HR = 1.8, P < 0.05). Immunohistochemistry showed that the positive rates of EZH2 and VEGF proteins in CRC tissues were significantly higher than those in adjacent tissues (P < 0.01). The positive expression of EZH2 was related to tumor diameter, differentiation, and Duke stage (P < 0.05). The positive expression of VEGF was correlated with degree of differentiation and Duke stage (P < 0.05).
CONCLUSION EZH2 and VEGF are up-regulated and mutated in CRC tissues. The high expression of EZH2 and VEGF is related to tumor size, differentiation, and Duke stage, and can be used as potential molecular markers for the prognosis of CRC.
Collapse
Affiliation(s)
- Li-Gang Yu
- Department of General Surgery, Tianjin Wuqing District People's Hospital, Tianjin 301700, China
| | - Su-Qin Chen
- Department of General Surgery, Tianjin Wuqing District People's Hospital, Tianjin 301700, China,Department of Pathology, Tianjin Wuqing District People's Hospital, Tianjin 301700, China
| | - Guo-Liang Mao
- Department of General Surgery, Tianjin Wuqing District People's Hospital, Tianjin 301700, China
| |
Collapse
|
43
|
Xu H, Hu M, Liu M, An S, Guan K, Wang M, Li L, Zhang J, Li J, Huang L. Nano-puerarin regulates tumor microenvironment and facilitates chemo- and immunotherapy in murine triple negative breast cancer model. Biomaterials 2020; 235:119769. [PMID: 31986348 PMCID: PMC7093100 DOI: 10.1016/j.biomaterials.2020.119769] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/22/2019] [Accepted: 01/07/2020] [Indexed: 01/05/2023]
Abstract
Tumor associated fibroblasts (TAFs) are key stromal cells mediating the desmoplastic reaction and being partially responsible for the drug-resistance and immunosuppressive microenvironment formation in solid tumors. Delivery of genotoxic drugs off-targetedly to kill TAFs results in production of Wnt16 which renders the neighboring tumor cells drug resistant as shown in our previous study (PMC4623876). Our current approach looks for means to deactivate, rather than kill, TAFs. Reactive oxygen species (ROS) are the central hub of multiple profibrogenic pathways and indispensable for TAFs activation. Herein, puerarin was identified to effectively downregulate ROS production in the activated myofibroblast. In this study, a novel puerarin nanoemulsion (nanoPue) was developed to improve the solubility and bioavailability of puerarin. NanoPue significantly deactivated the stromal microenvironment (e.g., ~6-fold reduction of TAFs in nanoPue treated mice compared with the PBS control, p < 0.0001) and facilitated chemotherapy effect of nano-paclitaxel in the desmoplastic triple-negative breast cancer (TNBC) model. Moreover, the removal of the physical barrier increased intra-tumoral infiltration of cytotoxic T cell by 2-fold. This activated immune microenvironment allowed nanoPue to synergize PD-L1 blockade therapy in TNBC model.
Collapse
Affiliation(s)
- Huan Xu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States; Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, People's Republic of China
| | - Mengying Hu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Mengrui Liu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Sai An
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Kaiyun Guan
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Menglin Wang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Lei Li
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Jing Zhang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States
| | - Jun Li
- ZY Therapeutics Inc., 400 Park Office Dr. Suite 310, Research Triangle Park, NC, 27709, United States
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States.
| |
Collapse
|
44
|
Liu Y, Guo J, Huang L. Modulation of tumor microenvironment for immunotherapy: focus on nanomaterial-based strategies. Am J Cancer Res 2020; 10:3099-3117. [PMID: 32194857 PMCID: PMC7053194 DOI: 10.7150/thno.42998] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 01/19/2020] [Indexed: 02/07/2023] Open
Abstract
Recent advances in the field of immunotherapy have profoundly opened up the potential for improved cancer therapy and reduced side effects. However, the tumor microenvironment (TME) is highly immunosuppressive, therefore, clinical outcomes of currently available cancer immunotherapy are still poor. Recently, nanomaterial-based strategies have been developed to modulate the TME for robust immunotherapeutic responses. In this review, the immunoregulatory cell types (cells relating to the regulation of immune responses) inside the TME in terms of stimulatory and suppressive roles are described, and the technologies used to identify and quantify these cells are provided. In addition, recent examples of nanomaterial-based cancer immunotherapy are discussed, with particular emphasis on those designed to overcome barriers caused by the complexity and diversity of TME.
Collapse
|
45
|
Relaxin-FOLFOX-IL-12 triple combination therapy engages memory response and achieves long-term survival in colorectal cancer liver metastasis. J Control Release 2019; 319:213-221. [PMID: 31899270 DOI: 10.1016/j.jconrel.2019.12.053] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/19/2019] [Accepted: 12/30/2019] [Indexed: 12/15/2022]
Abstract
Induction of memory T cell response is inefficient in colorectal cancer (CRC) liver metastasis following existing therapies due to abundant stroma and immunosuppressive environment within the metastatic liver, which leads to fast disease progression, high recurrence rate, and short survival. Two fundamental steps are involved to elicit extensive memory T cell response: stimulation of naive T cells with robust and persistent antigen signals; and maintenance of differentiated memory T cells with survival factors. Here, we demonstrate a rational design of triple combination regimen, including relaxin (RLN), FOLFOX (combination of 5-fluorouracil, leucovorin, and oxaliplatin), and IL-12, successfully stimulates central memory T cells and achieves long-term survival in an aggressive experimental CRC liver metastasis model. Sequential administration of FOLFOX and IL-12 gene therapy following stromal deactivation by RLN gene therapy completely cured established CRC liver metastases in ~50% of mice and provided long-lasting protection against tumor recurrence. The study here may highlight the potential of evoking memory response as a curative therapy for the treatment of CRC liver metastasis.
Collapse
|
46
|
Di Mascolo D, Varesano S, Benelli R, Mollica H, Salis A, Zocchi MR, Decuzzi P, Poggi A. Nanoformulated Zoledronic Acid Boosts the Vδ2 T Cell Immunotherapeutic Potential in Colorectal Cancer. Cancers (Basel) 2019; 12:E104. [PMID: 31906080 PMCID: PMC7017311 DOI: 10.3390/cancers12010104] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/19/2019] [Accepted: 12/29/2019] [Indexed: 12/11/2022] Open
Abstract
Aminobisphosphonates, such as zoledronic acid (ZA), have shown potential in the treatment of different malignancies, including colorectal carcinoma (CRC). Yet, their clinical exploitation is limited by their high bone affinity and modest bioavailability. Here, ZA is encapsulated into the aqueous core of spherical polymeric nanoparticles (SPNs), whose size and architecture resemble that of biological vesicles. On Vδ2 T cells, derived from the peripheral blood of healthy donors and CRC patients, ZA-SPNs induce proliferation and trigger activation up to three orders of magnitude more efficiently than soluble ZA. These activated Vδ2 T cells kill CRC cells and tumor spheroids, and are able to migrate toward CRC cells in a microfluidic system. Notably, ZA-SPNs can also stimulate the proliferation of Vδ2 T cells from the tumor-infiltrating lymphocytes of CRC patients and boost their cytotoxic activity against patients' autologous tumor organoids. These data represent a first step toward the use of nanoformulated ZA for immunotherapy in CRC patients.
Collapse
Affiliation(s)
- Daniele Di Mascolo
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, 16163 Genoa, Italy; (D.D.M.); (H.M.); (P.D.)
| | - Serena Varesano
- Molecular Oncology and Angiogenesis Unit, IRCCS Policlinico San Martino, 16132 Genoa, Italy;
| | - Roberto Benelli
- Immunology Unit, Ospedale Policlinico San Martino, University of Genoa, 16132 Genoa, Italy;
| | - Hilaria Mollica
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, 16163 Genoa, Italy; (D.D.M.); (H.M.); (P.D.)
| | | | - Maria Raffaella Zocchi
- Division of Immunology, Transplants and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, 16163 Genoa, Italy; (D.D.M.); (H.M.); (P.D.)
| | - Alessandro Poggi
- Molecular Oncology and Angiogenesis Unit, IRCCS Policlinico San Martino, 16132 Genoa, Italy;
| |
Collapse
|
47
|
Huang X, Lin H, Huang F, Xie Y, Wong KH, Chen X, Wu D, Lu A, Yang Z. Targeting Approaches of Nanomedicines in Acute Myeloid Leukemia. Dose Response 2019; 17:1559325819887048. [PMID: 31853234 PMCID: PMC6906351 DOI: 10.1177/1559325819887048] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 09/10/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy, which is commonly
associated with high incidence and mortality among adult patients. The standard
induction regimen for AML has been substantially unchanged over the past 40
years, for which novel nanomedicines have represented a promising strategy in
AML therapies. Despite developments of multiple nanoparticles formulated with
drugs or genes, less there is not much information available about approaches in
AML is available. This review presents an overview of nanomedicines currently
being evaluated in AML. First, it briefly summarized conventional chemotherapies
in use. Second, nanomedicines presently ongoing in clinical trials or
preclinical researches were classified and described, with illustrative examples
from recent literatures. Finally, limitations and potential safety issues
concerns in clinical translation of AML treatment were discussed as well.
Collapse
Affiliation(s)
- Xiao Huang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Hai Lin
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Feng Huang
- Institute of Acupuncture & Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuning Xie
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Ka Hong Wong
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Xiaoyu Chen
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Dongyue Wu
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Zhijun Yang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| |
Collapse
|
48
|
Chen F, Alphonse M, Liu Q. Strategies for nonviral nanoparticle-based delivery of CRISPR/Cas9 therapeutics. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 12:e1609. [PMID: 31797562 DOI: 10.1002/wnan.1609] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 11/08/2019] [Accepted: 11/12/2019] [Indexed: 12/26/2022]
Abstract
CRISPR-based genome editing technology has become an important potential therapeutic tool for various diseases. A vital challenge is to reach a safe, efficient, and clinically suitable delivery of a CRISPR-associated protein and a single-guide RNA. A possible translational approach to applying CRISPR-based technology is the use of viral vectors such as adeno-associated virus. However, such vectors give long-term exposure in vivo that may increase potential off-target effects as well as the risk of immunogenicity. Therefore, limitations to clinical applications are addressed using nonviral delivery systems such as nanoparticle-based delivery strategies. Today, the nanoparticle-based delivery approach is becoming more and more attractive in gene therapeutics because of its specific targeting, scale-up efficiency, efficacy of customization, minor stimulation of immune response, and minimal exposure to nucleases. In this review, we will present the most recent advances in developing innovations and potential advantages of the nanoparticle delivery system in CRISPR genome editing. We will also propose potential strategies of CRISPR-based technology for therapeutic and industrial applications. Our review will differ in focus from previous reviews and advance the literature on the subject by (a) focusing on the challenges of the CRISPR/Cas9 delivery system; (b) focusing on the application of nanoparticle-based delivery of CRISPR components (Cas9 and sgRNA), such as lipids and polymeric vectors; (c) discussing the potential nanoparticle-based delivery approaches for CRISPR/Cas9 application. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Fengqian Chen
- Department of Environmental Toxicology, The Institute of Environmental and Human Health (TIEHH), Texas Tech University, Lubbock, Texas
| | - Martin Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Qi Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
49
|
Cheng X, Huang Z, Long D, Jin W. BET inhibitor bromosporine enhances 5-FU effect in colorectal cancer cells. Biochem Biophys Res Commun 2019; 521:840-845. [PMID: 31708100 DOI: 10.1016/j.bbrc.2019.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 11/02/2019] [Indexed: 12/17/2022]
Abstract
Treatment of colorectal cancer (CRC) remains a challenge because of the lack of effective early treatment strategies and high incidence of relapse. 5-Fluorouracil (5-FU) is a typical CRC treatment. Bromosporine is an innovative bromodomain and extraterminal domain (BET) inhibitor. We investigated if CRC could be targeted by the combination of 5-FU and bromosporine in a synergistic manner in vivo and in vitro. Our findings shown that the combination treatment inhibits cell viability, formation of colonies, increased apoptosis and cell cycle arrest at G0-G1. In addition, the expression level of BRD4 was high in HCT116 cells exposed to 5-FU that showed lower apoptosis against the parental cells. Moreover, the 5-FU-resistance was reversed significantly by BRD4 knockdown or inhibition. The drug combination showed increased activity against tumor than individual drug exposure in the xenograft model. In conclusion, this work serves as a basic clinical evaluation of 5-FU and bromosporine as an effective therapeutic approach for CRC.
Collapse
Affiliation(s)
- Xueyuan Cheng
- Department of General Surgery, The Ninth Affiliated Hospital of Guangxi Medical University, Beihai People's Hospital, Beihai, 536000, Guangxi Zhuang, China
| | - Zhong Huang
- Department of General Surgery, The Ninth Affiliated Hospital of Guangxi Medical University, Beihai People's Hospital, Beihai, 536000, Guangxi Zhuang, China
| | - Di Long
- Department of General Surgery, Wuming Hospital of Guangxi Medical University, Nanning, 530199, Guangxi Zhuang, China.
| | - Wei Jin
- Department of General Surgery, Wuming Hospital of Guangxi Medical University, Nanning, 530199, Guangxi Zhuang, China
| |
Collapse
|