51
|
Wang P, Chen W, Zhang Y, Zhong Q, Li Z, Wang Y. MicroRNA-1246 suppresses the metastasis of breast cancer cells by targeting the DYRK1A/PGRN axis to prevent the epithelial-mesenchymal transition. Mol Biol Rep 2022; 49:2711-2721. [PMID: 35059968 DOI: 10.1007/s11033-021-07080-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 12/09/2021] [Indexed: 10/19/2022]
Abstract
OBJECTIVE Breast cancer is one of the most common malignant and highly heterogeneous tumors in women. MicroRNAs (miRNAs), such as miR-1246, play important roles in various types of malignant cancers, including triple-negative breast cancer (TNBC). However, the biological role of miR-1246 in TNBC has not yet been fully elucidated. In this study, we studied the role of miR-1246 in the occurrence and development of TNBC and its mechanism of action. METHODS Cell Counting Kit-8 (CCK-8), wound healing, and Transwell assays were performed to observe the effects of miR-1246 on TNBC cell proliferation, migration, and invasion, respectively. The expression of epithelial-mesenchymal transition (EMT) markers was detected by western blotting. Dual luciferase reporter assays were performed to determine whether DYRK1A is a novel target of miR-1246. In addition, an immunoprecipitation experiment was performed to verify the binding of DYRK1A to PGRN. Rescue experiments were performed to determine whether DYRK1A is a novel target of miR-1246 and whether miR-1246 suppresses the metastasis of breast cancer cells by targeting the DYRK1A/PGRN axis to prevent the epithelial-mesenchymal transition. RESULTS Our results show that miR‑1246 suppresses the proliferation, migration, and invasion of TNBC cells, DYRK1A is a novel target of miR-1246 and Importin-8 mediated miR-1246 nuclear translocation. MiR‑1246 plays a suppressive role in the regulation of the EMT of TNBC cells by targeting DYRK1A. DYRK1A mediates the metastasis of triple-negative breast cancer via activation of the EMT. We identified PGRN as a novel DYRK1A-interacting protein. Overexpression of PGRN and DYRK1A promoted cell proliferation and migration of TNBC, but this effect was reversed by co-expression of miR-1246 mimics.DYRK1A and PGRN act together to regulate the occurrence and development of breast cancer through miR-1246. CONCLUSION MiR-1246 suppresses the metastasis of breast cancer cells by targeting the DYRK1A/PGRN axis and preventing the epithelial-mesenchymal transition. The MiR-1246/DYRK1A/PGRN axis regulates TNBC progression, suggesting that MiR-1246 could be promising therapeutic targets for the treatment of TNBC.
Collapse
Affiliation(s)
- Pan Wang
- Department of Laboratory Medicine, Taizhou Central Hospital, Taizhou, 318000, Zhejiang, China
| | - Wenju Chen
- Department of Laboratory Medicine, Taizhou Central Hospital, Taizhou, 318000, Zhejiang, China
| | - Yaqiong Zhang
- Department of Laboratory Medicine, Taizhou Central Hospital, Taizhou, 318000, Zhejiang, China
| | - Qianyi Zhong
- Department of Laboratory Medicine, Taizhou Central Hospital, Taizhou, 318000, Zhejiang, China
| | - Zhaoyun Li
- Department of Laboratory Medicine, Taizhou Central Hospital, Taizhou, 318000, Zhejiang, China
| | - Yichao Wang
- Department of Laboratory Medicine, Taizhou Central Hospital, Taizhou, 318000, Zhejiang, China.
- Department of Clinical Laboratory Medicine, Taizhou Central Hospital (Taizhou University Hospital), No. 999 Donghai Road, Jiaojiang District, Taizhou, 318000, Zhejiang, People's Republic of China.
| |
Collapse
|
52
|
Lin C, Chen YZ, Wu B, Yang MT, Liu CQ, Zhao Y. Advances and prospects of ultrasound targeted drug delivery systems using biomaterial-modified micro/nanobubbles for tumor therapy. Curr Med Chem 2022; 29:5062-5075. [PMID: 35362371 DOI: 10.2174/0929867329666220331110315] [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: 10/04/2021] [Revised: 01/18/2022] [Accepted: 02/01/2022] [Indexed: 11/22/2022]
Abstract
The incidence of malignant tumors is rising rapidly and tends to be in the younger, which has been one of the most important factors endangering the safety of human life. Ultrasound micro/nanobubbles, as a noninvasive and highly specific antitumor strategy, can reach and destroy tumor tissue through their effects of cavitation and acoustic perforation under the guidance of ultrasound. Meanwhile, micro/nanobubbles are now used as a novel drug carrier, releasing drugs at a target region, especially on the prospects of biomaterial-modified micro/nanobubbles as a dual modality for drug delivery and therapeutic monitoring. and successful evaluation of the sonoporation mechanism(s), ultrasound parameters, drug type and dose will need to be addressed before translating this technology for clinical use. Therefore, this paper collects the literature on the experimental and clinical studies of ultrasound biomaterial-modified micro/nanobubbles therapy in vitro and in vivo in recent years.
Collapse
Affiliation(s)
- Chen Lin
- Medical College of China three Gorges University;Yichang; China
| | - Ye-Zi Chen
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy,China Three Gorges University; Yichang; China
| | - Bo Wu
- Medical College of China three Gorges University;Yichang; China
| | - Meng-Ting Yang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy,China Three Gorges University; Yichang; China
| | - Chao-Qi Liu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy,China Three Gorges University; Yichang; China
| | - Yun Zhao
- Medical College of China three Gorges University;Yichang; China
| |
Collapse
|
53
|
Wang C, Li SQ, Zhao Y. Effects of different administration strategies of cetuximab on remission rate, MMPs, miR-106b-5p, PCAT-1, and adverse reactions in patients with RAS wild-type CRC liver metastasis. Shijie Huaren Xiaohua Zazhi 2022; 30:204-211. [DOI: 10.11569/wcjd.v30.i4.204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) has no obvious clinical symptoms at the initial stage of the disease, and it is often diagnosed at an advanced stage of pathology, with distant metastasis. The most common target organ of distant metastasis in CRC is the liver, and patients with liver metastasis have been shown to be well treated with biologic targeted agents in combination with FOLFIRI regimen.
AIM To evaluate the efficacy and safety of different administ-ration strategies of cetuximab in patients with RAS wild-type CRC with liver metastasis.
METHODS From July 2017 to March 2019, 74 patients with RAS wild-type CRC liver metastases treated with the FOLFOX regimen at our hospital were selected as the research subjects. According to the cetuximab administration strategy used, they were divided into either a weekly group or a bi-weekly group, with 37 cases in each group. The weekly group was initially given 400 mg/m2 cetuximab by intravenous infusion over 120 min, followed by 250 mg/m2 cetuximab once every week. The bi-weekly group was given 500 mg/m2 cetuximab once every 2 wk. The disease remission rate, the incidence of toxic and side effects, the improvement of quality of life, the survival, and the expression of matrix metalloproteinases (MMPs), miR-106b-5p, and prostate cancer-related non-coding RNA transcript 1 (PCAT-1) before and after treatment were compared between the two groups.
RESULTS The disease remission rate in the biweekly group was significantly higher than that of the weekly group (78.38% vs 54.05%, P < 0.05). The incidence of leukopenia and/or neutropenia, oral mucositis, and acne-like rash in the biweekly group was significantly higher than that of the weekly group (P < 0.05). The relative expression of MMP-2 and MMP-9 in the biweekly group was significantly lower than that in the weekly group, but the expression of tissue inhibitor of matrix metalloproteinases (T1MP) was significantly higher than that in the weekly group (P < 0.05). The relative expression of miR-106b-5p and PCAT-1 after treatment was significantly lower in both groups, and the decrease was significantly greater in the bi-weekly group (P < 0.05). The effective rate of improving the quality of life was significantly higher in the bi-weekly group than in the weekly group (72.97% vs 48.65%, P < 0.05), though there was no statistically significant difference in the progression-free survival time between the two groups (P > 0.05).
CONCLUSION Cetuximab biweekly treatment of RAS wild-type CRC patients with liver metastases has better remission rate and quality of life improvement than weekly administration, but it increases the incidence of adverse reactions in patients. The administration strategy can be selected according to the patient's tolerance.
Collapse
Affiliation(s)
- Chao Wang
- Clinical Laboratory, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
| | - Sheng-Qiang Li
- Clinical Laboratory, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
| | - Yan Zhao
- Clinical Laboratory, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
| |
Collapse
|
54
|
Chen Y, Yang M, Meng F, Zhang Y, Wang M, Guo X, Yang J, Zhang H, Zhang H, Sun J, Wang W. SRSF3 Promotes Angiogenesis in Colorectal Cancer by Splicing SRF. Front Oncol 2022; 12:810610. [PMID: 35198444 PMCID: PMC8859257 DOI: 10.3389/fonc.2022.810610] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/17/2022] [Indexed: 12/14/2022] Open
Abstract
SRSF3, an important member of the serine/arginine-rich protein (SRp) family, is highly expressed in various tumors and plays an important role in tumor cell proliferation, migration and invasion. However, it is still unclear whether SRSF3 is involved in tumor angiogenesis. In this study, we first revealed that SRSF3 regulated the expression of numerous genes related to angiogenesis, including proangiogenic SRF. Then, we confirmed that SRSF3 was highly expressed in colorectal cancer (CRC) and was positively correlated with SRF. Mechanistic studies revealed that SRSF3 directly bound to the “CAUC” motif in exon 6 of SRF and induced the exclusion of introns. Knockdown of SRSF3 significantly reduced the secretion of VEGF from CRC cells. Conditioned medium from SRSF3-knockdown CRC cells significantly inhibited the migration, invasion and tube formation of human umbilical vein endothelial cells (HUVECs). In addition, SRF silencing inhibited angiogenesis, while SRF overexpression reversed the antiangiogenic effects of SRSF3 knockdown on tube formation. These findings indicate that SRSF3 is involved in the splicing of SRF and thereby regulates the angiogenesis of CRC, which offers novel insight into antiangiogenic therapy in CRC.
Collapse
Affiliation(s)
- Yinshuang Chen
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Man Yang
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Fanyi Meng
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yawen Zhang
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Mengmeng Wang
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Xuqin Guo
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jie Yang
- Institute of Medical Technology, Suzhou Vocational Health College, Suzhou, China
| | - Hongjian Zhang
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Haiyang Zhang
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jing Sun
- Institute of Medical Technology, Suzhou Vocational Health College, Suzhou, China
| | - Weipeng Wang
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
55
|
Liu B, Yao C, Li H. Laparoscopic Radical Resection of Colorectal Cancer in the Treatment of Elderly Colorectal Cancer and Its Effect on Gastrointestinal Function. Front Surg 2022; 9:840461. [PMID: 35284487 PMCID: PMC8907596 DOI: 10.3389/fsurg.2022.840461] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/06/2022] [Indexed: 12/01/2022] Open
Abstract
Objective To explore the efficacy and safety of laparoscopic radical resection of colorectal cancer in the elderly patients and its impact on gastrointestinal function. Methods A total of 122 elderly patients with colorectal cancer admitted to our hospital from March 2020 to June 2021 were selected as the research subjects, and they were divided into the control group (n = 61) and the observation group (n = 61). The control group was treated with traditional laparotomy, and the observation group was treated with laparoscopic radical resection of colorectal cancer. The clinical data of operation time, incision length, intraoperative bleeding volume, and hospitalization time in the two groups were recorded. Serum motilin (MTL) and gastrin (GAS) levels were measured pre- and post-operatively. The duration of abdominal distension, the time for the abdominal sound to return to normal, the time for the anal exhaust to normal, and the time for normal food intake were recorded after operation. The patients were followed up for 6 months post-operatively, and the complications during follow-up were recorded. Results The total response rate of the observation group (95.08%) was higher than that of the control group (81.97%) (P < 0.05). The operation time, incision length, intraoperative bleeding volume, and hospitalization time of the observation group were lower than those of the control group (P < 0.05). The duration of abdominal distension, the time for bowel sounds to return to normal, the time for the anus to exhaust gas to normal, and the normal eating time in the observation group were all lower than those in the control group (P < 0.05). After surgery, the levels of MTL and GAS in the two groups were lower than those before surgery, and those in the observation group were lower than those in the control group (P < 0.05). The total incidence of complications in the observation group (3.28%) was lower than that in the control group (13.12%) (P < 0.05). Conclusion Laparoscopic radical resection of colorectal cancer in the elderly patients has good effect, short operation time, less trauma, less blood loss during operation, short hospital stay, good recovery of gastrointestinal function, fewer complications, and high safety.
Collapse
Affiliation(s)
- Biao Liu
- The Third Department of Surgery, Cangxian Hospital, Cangzhou, China
- *Correspondence: Biao Liu
| | - Chuanhui Yao
- The First Department of Surgery, Cangxian Hospital, Cangzhou, China
| | - Haiying Li
- Department of Medical Affairs, Cangxian Hospital, Cangzhou, China
| |
Collapse
|
56
|
Immunotherapy for Colorectal Cancer: Mechanisms and Predictive Biomarkers. Cancers (Basel) 2022; 14:cancers14041028. [PMID: 35205776 PMCID: PMC8869923 DOI: 10.3390/cancers14041028] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Late-stage colorectal cancer treatment often involves chemotherapy and radiation that can cause dose-limiting toxicity, and therefore there is great interest in developing targeted therapies for this disease. Immunotherapy is a targeted therapy that uses peptides, cells, antibodies, viruses, or small molecules to engage or train the immune system to kill cancer. Here, we discuss the preclinical and clinical development of immunotherapy for treatment of colorectal cancer and provide an overview of predictive biomarkers for such treatments. We also consider open questions including optimal combination treatments and sensitization of colorectal cancer patients with proficient mismatch repair enzymes. Abstract Though early-stage colorectal cancer has a high 5 year survival rate of 65–92% depending on the specific stage, this probability drops to 13% after the cancer metastasizes. Frontline treatments for colorectal cancer such as chemotherapy and radiation often produce dose-limiting toxicities in patients and acquired resistance in cancer cells. Additional targeted treatments are needed to improve patient outcomes and quality of life. Immunotherapy involves treatment with peptides, cells, antibodies, viruses, or small molecules to engage or train the immune system to kill cancer cells. Preclinical and clinical investigations of immunotherapy for treatment of colorectal cancer including immune checkpoint blockade, adoptive cell therapy, monoclonal antibodies, oncolytic viruses, anti-cancer vaccines, and immune system modulators have been promising, but demonstrate limitations for patients with proficient mismatch repair enzymes. In this review, we discuss preclinical and clinical studies investigating immunotherapy for treatment of colorectal cancer and predictive biomarkers for response to these treatments. We also consider open questions including optimal combination treatments to maximize efficacy, minimize toxicity, and prevent acquired resistance and approaches to sensitize mismatch repair-proficient patients to immunotherapy.
Collapse
|
57
|
Sousa MJD, Gomes I, Pereira TC, Magalhães J, Basto R, Paulo J, Jacinto P, Bonito N, Sousa G. The effect of prognostic factors at baseline on the efficacy of trifluridine/tipiracil in patients with metastatic colorectal cancer: A Portuguese exploratory analysis. Cancer Treat Res Commun 2022; 31:100531. [PMID: 35172243 DOI: 10.1016/j.ctarc.2022.100531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/03/2022] [Accepted: 02/04/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND The RECOURSE trial supported trifluridine/tipiracil as a treatment option in metastatic colorectal cancer (mCRC). Subsequent analysis demonstrated that low tumour burden and indolent disease are good prognosis factors improving progression-free survival (PFS) and overall survival (OS). This study aimed to evaluate the impact of prognosis group in the OS, PFS and safety of trifluridine/tipiracil in patients with mCRC. METHODS Single-centre, retrospective, and observational study of patients with mCRC who started trifluridine/tipiracil between February 2018 and July 2019. Patients were divided into good prognosis characteristics (GPC) [low tumour burden (less than 3 metastasis site) and indolent disease (≥18 months from first metastasis diagnosis)] and poor prognostic characteristics (PPC) group [high tumour burden (3 or more metastasis sites) and/or aggressive disease (<18 months since the first metastasis diagnosis)]. RESULTS Median age was 67 years (48-82), 67.3% of the patients were male, and 65.3% had stage IV disease at baseline. Overall, median OS was 7.5 months (95%CI:5.7-9.3). Twenty-two patients (44.9%) presented GPC and 29 (59.1%) had PPC. GPC patients had longer median OS [11.4 (95%CI:6.2-16.7)] versus 3.9 months [(95%CI: 3.3-4.6),p < 0.0001] and PFS [4.9 (95%CI:3.0-6.9) versus 2.6 months (95%CI:2.2-2.8),p < 0.0001]. These differences were more pronounced in GPC patients with no liver metastasis. Grade ≥3 adverse events incidence didn't vary between GPC and PPC subgroups. CONCLUSION Our study validates the improved trifluridine/tipiracil efficacy in patients with GPC in comparison with PPC while maintaining a well-tolerated safety profile. Indolent disease, low tumour burden and the absence of liver metastasis were shown to be good prognosis factors influencing sustained response to trifluridine/tipiracil.
Collapse
Affiliation(s)
- M João de Sousa
- Medical Oncology Service, Institute of Oncology Francisco Gentil (IPO Coimbra), Coimbra, Portugal.
| | - Inês Gomes
- Medical Oncology Service, Institute of Oncology Francisco Gentil (IPO Coimbra), Coimbra, Portugal
| | - Tatiana Cunha Pereira
- Medical Oncology Service, Institute of Oncology Francisco Gentil (IPO Coimbra), Coimbra, Portugal
| | - Joana Magalhães
- Medical Oncology Service, Institute of Oncology Francisco Gentil (IPO Coimbra), Coimbra, Portugal
| | - Raquel Basto
- Medical Oncology Service, Institute of Oncology Francisco Gentil (IPO Coimbra), Coimbra, Portugal
| | - Judy Paulo
- Medical Oncology Service, Institute of Oncology Francisco Gentil (IPO Coimbra), Coimbra, Portugal
| | - Paula Jacinto
- Medical Oncology Service, Institute of Oncology Francisco Gentil (IPO Coimbra), Coimbra, Portugal
| | - Nuno Bonito
- Medical Oncology Service, Institute of Oncology Francisco Gentil (IPO Coimbra), Coimbra, Portugal
| | - Gabriela Sousa
- Medical Oncology Service, Institute of Oncology Francisco Gentil (IPO Coimbra), Coimbra, Portugal
| |
Collapse
|
58
|
Leung WH, Shih JW, Chen JS, Mokgautsi N, Wei PL, Huang YJ. Preclinical Identification of Sulfasalazine's Therapeutic Potential for Suppressing Colorectal Cancer Stemness and Metastasis through Targeting KRAS/MMP7/CD44 Signaling. Biomedicines 2022; 10:377. [PMID: 35203586 PMCID: PMC8962339 DOI: 10.3390/biomedicines10020377] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/27/2022] [Accepted: 01/29/2022] [Indexed: 02/04/2023] Open
Abstract
Approximately 25% of colorectal cancer (CRC) patients will develop metastatic (m)CRC despite treatment interventions. In this setting, tumor cells are attracted to the epidermal growth factor receptor (EGFR) oncogene. Kirsten rat sarcoma (RAS) 2 viral oncogene homolog (KRAS) mutations were reported to drive CRC by promoting cancer progression in activating Wnt/β-catenin and RAS/extracellular signal-regulated kinase (ERK) pathways. In addition, KRAS is associated with almost 40% of patients who acquire resistance to EGFR inhibitors in mCRC. Multiple studies have demonstrated that cancer stem cells (CSCs) promote tumorigenesis, tumor growth, and resistance to therapy. One of the most common CSC prognostic markers widely reported in CRC is a cluster of differentiation 44 (CD44), which regulates matrix metalloproteinases 7/9 (MMP7/9) to promote tumor progression and metastasis; however, the molecular role of CD44 in CRC is still unclear. In invasive CRC, overexpression of MMP7 was reported in tumor cells compared to normal cells and plays a crucial function in CRC cetuximab and oxaliplatin resistance and distant metastasis. Here, we utilized a bioinformatics analysis and identified overexpression of KRAS/MMP7/CD44 oncogenic signatures in CRC tumor tissues compared to normal tissues. In addition, a high incidence of mutations in KRAS and CD44 were associated with some of the top tumorigenic oncogene's overexpression, which ultimately promoted a poor response to chemotherapy and resistance to some FDA-approved drugs. Based on these findings, we explored a computational approach to drug repurposing of the drug, sulfasalazine, and our in silico molecular docking revealed unique interactions of sulfasalazine with the KRAS/MMP7/CD44 oncogenes, resulting in high binding affinities compared to those of standard inhibitors. Our in vitro analysis demonstrated that sulfasalazine combined with cisplatin reduced cell viability, colony, and sphere formation in CRC cell lines. In addition, sulfasalazine alone and combined with cisplatin suppressed the expression of KRAS/MMP7/CD44 in DLD-1 and HCT116 cell lines. Thus, sulfasalazine is worthy of further investigation as an adjuvant agent for improving chemotherapeutic responses in CRC patients.
Collapse
Affiliation(s)
- Wai-Hung Leung
- Division of Colon and Rectal Surgery, Department of Surgery, Mackay Memorial Hospital, No. 92, Sec. 2, Zhongshan N. Rd., Taipei 10449, Taiwan; (W.-H.L.); (J.-S.C.)
| | - Jing-Wen Shih
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan; (J.-W.S.); (N.M.)
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Jian-Syun Chen
- Division of Colon and Rectal Surgery, Department of Surgery, Mackay Memorial Hospital, No. 92, Sec. 2, Zhongshan N. Rd., Taipei 10449, Taiwan; (W.-H.L.); (J.-S.C.)
| | - Ntlotlang Mokgautsi
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan; (J.-W.S.); (N.M.)
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Po-Li Wei
- Division of Colorectal Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei Medical University, Taipei 110, Taiwan;
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei Medical University, Taipei 110, Taiwan
| | - Yan-Jiun Huang
- Division of Colorectal Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei Medical University, Taipei 110, Taiwan;
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
59
|
Yue Y, Zhang Q, Sun Z. CX3CR1 Acts as a Protective Biomarker in the Tumor Microenvironment of Colorectal Cancer. Front Immunol 2022; 12:758040. [PMID: 35140706 PMCID: PMC8818863 DOI: 10.3389/fimmu.2021.758040] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/28/2021] [Indexed: 12/12/2022] Open
Abstract
The tumor microenvironment (TME) plays an important role in the pathogenesis of many cancers. We aimed to screen the TME-related hub genes of colorectal adenoma (CRAD) and identify possible prognostic biomarkers. The gene expression profiles and clinical data of 464 CRAD patients in The Cancer Genome Atlas (TCGA) database were downloaded. The Estimation of STromal and Immune cells in MAlignant Tumours using Expression data (ESTIMATE) algorithm was performed to calculate the ImmuneScore, StromalScore, and EstimateScore. Thereafter, differentially expressed genes (DEGs) were screened. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, and protein–protein interaction (PPI) analysis were performed to explore the roles of DEGs. Furthermore, univariate and multivariate Cox analyses were accomplished to identify independent prognostic factors of CRAD. CX3CR1 was selected as a hub gene, and the expression was confirmed in colorectal cancer (CRC) patients and cell lines. The correlations between CX3CR1 and tumor-infiltrating immune cells were estimated by Tumor IMmune Estimation Resource database (TIMER) and CIBERSORT analysis. Besides, we investigated the effects of coculture with THP-1-derived macrophages with HCT8 cells with low CX3CR1 expression on immune marker expression, cell viability, and migration. There were significant differences in the ImmuneScore and EstimateScore among different stages. Patients with low scores presented significantly lower lifetimes than those in the high-score group. Moreover, we recognized 1,578 intersection genes in ImmuneScore and StromalScore, and these genes were mainly enriched in numerous immune-related biological processes. CX3CR1 was found to be associated with immune cell infiltration levels, immune marker expression, and macrophage polarization. Simultaneous silencing of CX3CR1 and coculture with THP-1 cells further regulated macrophage polarization and promoted the cell proliferation and migration of CRC cells. CX3CR1 was decreased in CRAD tissues and cell lines and was related to T and N stages, tumor differentiation, and prognosis. Our results suggest that CX3CR1 contributes to the recruitment and regulation of immune-infiltrating cells and macrophage polarization in CRC and TAM-induced CRC progression. CX3CR1 may act as a prognostic biomarker in CRC.
Collapse
Affiliation(s)
- Yuanyi Yue
- Department of Gastroenterology Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qiang Zhang
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhengrong Sun
- BioBank, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Zhengrong Sun,
| |
Collapse
|
60
|
Jin C, Zhu X, Huang X, Gong T, Wei Z, You J. Efficacy and safety of PD-1/PD-L1 and CTLA-4 immune checkpoint inhibitors in colorectal cancer: a meta-analysis. J Comp Eff Res 2022; 11:203-212. [PMID: 35023361 DOI: 10.2217/cer-2021-0134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Aims: To evaluate the efficacy and safety of PD-1/PD-L1 and/or CTLA-4 inhibitors in the treatment of colorectal cancer (CRC) by meta-analysis. Methods: Electronic databases were searched. Eligible studies included investigations of efficacy and safety of anti-PD-1/PD-L1 or anti-CTLA-4 agents in patients with CRC. Corresponding indicators were calculated. Results: A total of 15 articles were included. The pooled objective response rate, overall survival rate, progression-free survival rate and adverse event rate were 33, 56, 46 and 59%, respectively. The objective response rates for CRC with deficient mismatch repair and CRC with proficient mismatch repair were 43 and 3%, respectively, in patients treated with PD-1 inhibitors. Conclusion: The authors' study indicates that PD-1/PD-L1 inhibitors manifest promising clinical responses in the treatment of CRC with deficient mismatch repair with acceptable treatment-related adverse events.
Collapse
Affiliation(s)
- Chunhui Jin
- Department of Oncology, Wuxi Hospital, Nanjing University of Chinese Medicine, Wuxi, 214001, China
| | - Xiaodan Zhu
- Department of Oncology, Wuxi Hospital, Nanjing University of Chinese Medicine, Wuxi, 214001, China
| | - Xiaona Huang
- Department of Oncology, Wuxi Hospital, Nanjing University of Chinese Medicine, Wuxi, 214001, China
| | - Tingjie Gong
- Department of Oncology, Wuxi Hospital, Nanjing University of Chinese Medicine, Wuxi, 214001, China
| | - Zhipeng Wei
- Department of Oncology, Wuxi Hospital, Nanjing University of Chinese Medicine, Wuxi, 214001, China
| | - Jianliang You
- Department of Oncology, Wuxi Hospital, Nanjing University of Chinese Medicine, Wuxi, 214001, China
| |
Collapse
|
61
|
Wu L, Wang W, Tian J, Qi C, Cai Z, Yan W, Xuan S, Shang A. Combination therapy with Nab-paclitaxel and the interleukin-15 fused with anti-human serum albumin nanobody as a synergistic treatment for colorectal cancer. Bioengineered 2022; 13:1942-1951. [PMID: 35019820 PMCID: PMC8805949 DOI: 10.1080/21655979.2021.2023997] [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: 11/03/2022] Open
Abstract
This study determines the effect of Nab-paclitaxel in combination with IL-15 fusion protein, containing IL-15 and an anti-HSA nanobody domain, on colorectal cancer bearing mice. In vitro binding test of IL15 fusion protein to HSA and Nab-paclitaxel, as well as CTLL-2 cell stimulation assay were performed. The tumor inhibitory effects of Nab-paclitaxel in combination with IL-15 fusion protein was evaluated in the HCT116 bearing murine model. Moreover, the population and function of cytotoxic T cells and M1 macrophages, as well as MDSCs and Treg cells, were also further examined. As a result, combination therapy of Nab-paclitaxel and IL-15 fusion protein effectively inhibits the tumor growth and produced a 78% reduction in tumor size for HCT116, as compared to vehicle group. In the TDLN for the combination group, there were 18% of CD8+ IFN-γ + T-cells and 0.47% CD4+CD25+FOXP3+ regulatory T-cells, as opposed to 5.0% and 5.1%, respectively, for the model control group. Combination therapy further exhibited enhanced suppressive effects on the accumulation of CD11b+GR-1+ MDSC in spleen and bone marrow. Furthermore, Nab-paclitaxel and IL-15 fusion protein showed a significant suppression of NF-κB-mediated immune suppressive markers and increased expression of CD8, Granzyme B, CD62L, CD49b, and CD86 without obvious organ toxicity. In conclusion, combination therapy of Nab-paclitaxel and IL-15 fusion protein can effectively stimulate the antitumor activity of immune effector cells, thereby inhibiting immunosuppressive cells within the TME of colorectal cancer, and the overall therapeutic effect has a significant advantage over monotherapy.AbbreviationsInterleukin 15, IL-15; Human serum albumin, HSA; Myeloid-derived suppressor cells, MDSC; Albumin binding domain, ABD; Tumor drainage lymph node, TDLN; Natural killer (NK); Tumor-draining lymph node (TDLN); Tumor infiltrating lymphocyte, TIL; Immunogenic cell death, ICD; Enhanced permeability retention, EPR; Liposomal doxorubicin, Doxil; 5-fluorouracil, 5-FU.
Collapse
Affiliation(s)
- Lipei Wu
- Department of Laboratory Medicine, Dongtai People's Hospital & Dongtai Hospital of Nantong University, Yancheng, China.,Department of Laboratory Medicine, Shanghai Tongji Hospital, Shanghai, China
| | - Weiwei Wang
- Department of Pathology, Tinghu People's Hospital, Yancheng, China
| | - Jiale Tian
- Department of Laboratory Medicine, Shanghai Tongji Hospital, Shanghai, China
| | - Chunrun Qi
- Department of Pathology, Tinghu People's Hospital, Yancheng, China
| | - Zhengxin Cai
- Department of Laboratory Medicine, Tinghu People's Hospital of Yancheng City, Yancheng, China
| | - Wenhui Yan
- Department of Laboratory Medicine, Tinghu People's Hospital of Yancheng City, Yancheng, China
| | - Shihai Xuan
- Department of Laboratory Medicine, Dongtai People's Hospital & Dongtai Hospital of Nantong University, Yancheng, China
| | - Anquan Shang
- Department of Laboratory Medicine, Shanghai Tongji Hospital, Shanghai, China
| |
Collapse
|
62
|
Li S, Ding J, Wang Y, Wang X, Lv L. CD155/TIGIT signaling regulates the effector function of tumor-infiltrating CD8+ T cell by NF-κB pathway in colorectal cancer. J Gastroenterol Hepatol 2022; 37:154-163. [PMID: 34734434 DOI: 10.1111/jgh.15730] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 10/23/2021] [Accepted: 10/28/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIM CD155/T-cell immunoglobulin and ITIM domain (TIGIT) suppressed anti-cancer immunity in several cancers, but its roles in colorectal cancer (CRC) were not clear. Here, we investigated its roles in CRC. METHODS The percentages of CD8+ T cells expressing TIGIT and secreting cytokines (IL-2, TNF-α, and IFNγ) were evaluated by flow cytometry. The expression level of CD155 was determined by western blot and immunohistochemistry. The levels of cytokines were determined by enzyme-linked immunosorbent assay. The activation of the nuclear factor-kappa B (NF-κB) pathway was examined by western blot and immunofluorescent assay. RESULTS T-cell immunoglobulin and ITIM domain was overexpressed on CD8+ T cells of CRC patients and mice. CD155 was overexpressed in mice CRC tissues and cells. The addition of CD155 recombinant protein could decrease the percentages of CD8+ T cells secreting cytokines. Blocking TIGIT could increase the percentages of cytokine-secreting CD8+ T cells. Coculturing with CD155-knockdown CRC cells could upregulate the percentages of CD8+ T cells secreting cytokines. Blocking TIGIT partially counteracted the effect of the knockdown of CD155. Besides, coculturing with CD155-knockdown CRC cells could promote the secretion of cytokines, activate the NF-κB pathway, and enhance the nuclear translocation of p65. And these effects were counteracted by the application of an NF-κB inhibitor. Finally, blocking TIGIT played anti-cancer roles such as suppression of tumor growth, increasing the percentages of cytokine-secreting CD8+ T cells and activation of the NF-κB signaling pathway. CONCLUSION Suppressing CD155/TIGIT exerted anti-cancer effects against CRC, and our findings provided a potential therapeutic approach to treat CRC.
Collapse
Affiliation(s)
- Shanshan Li
- Department of Oncology, Zoucheng People's Hospital, Jining City, China
| | - Jihu Ding
- Department of Oncology, Shandong Chengwu County People's Hospital, Heze City, China
| | - Ying Wang
- Ward of TCM Anorectal Surgery, Feicheng Hospital Affiliated to Shandong First Medical University, Taian City, China
| | - Xianling Wang
- The First Department of Digestive Endoscopy, The Fourth Affiliated Hospital of China Medical University, Shenyang City, China
| | - Lin Lv
- Department of Health Care, Jinan Municipal Hospital of Traditional Chinese Medicine, Jinan City, China
| |
Collapse
|
63
|
Chi C, Liu T, Yang S, Wang B, Han W, Li J. ISLR affects colon cancer progression by regulating the epithelial-mesenchymal transition signaling pathway. Anticancer Drugs 2022; 33:e670-e679. [PMID: 34520435 PMCID: PMC8670340 DOI: 10.1097/cad.0000000000001233] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 07/28/2021] [Indexed: 11/27/2022]
Abstract
This study aims to determine the mechanism of ISLR on the progression of colon cancer. TCGA database was used to analyze ISLR expression in colon cancer tumor tissues. QRT-PCR and western blotting were used to detect ISLR expression in colon cancer cells. CCK-8, colony formation, EDU, wound healing and transwell assays were used to measure cell viability, proliferation, migration and invasion of colon cancer cells, respectively. The signaling pathway enrichment analysis of ISLR was analyzed on the basis of the KEGG database. The protein expression of genes related to signaling pathway was measured by western blotting. Results of TCGA analysis, qRT-PC and western blotting showed that ISLR was upregulated in colon cancer tumor tissues and cells. High level of ISLR was related to low overall survival of patients with colon cancer. ISLR silence significantly inhibited cell viability, proliferation, migration and invasion of colon cancer cells. ISLR overexpression markedly enhanced the cell viability, proliferation, migration and invasion of colon cancer cells. KEGG database analyzed showed that ISLR can activate the EMT signaling pathway. Inhibition of the EMT signaling pathway can suppress the growth, migration, and invasion of colon cancer cells and eliminate the promoted effect of ISLR overexpression on colon cancer progression. ISLR promotes the progression of colon cancer by activating the EMT signaling pathway.
Collapse
Affiliation(s)
- Chunhua Chi
- Department of Anorectal Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan
| | - Tongming Liu
- Department of Anorectal Surgery, Feicheng People’s hospital, Tai An
| | - Shengnan Yang
- Department of Proctology, Changqing District Hospital of Traditional Chinese Medicine, Jinan, Shandong, P.R. China
| | - Benjun Wang
- Department of Anorectal Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan
| | - Weiwei Han
- Department of Anorectal Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan
| | - Jiansheng Li
- Department of Anorectal Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan
| |
Collapse
|
64
|
Li T, Yan Z, Wang W, Zhang R, Gan W, Lv S, Zeng Z, Hou Y, Yang M. SEMA6B Overexpression Predicts Poor Prognosis and Correlates With the Tumor Immunosuppressive Microenvironment in Colorectal Cancer. Front Mol Biosci 2021; 8:687319. [PMID: 34938771 PMCID: PMC8687481 DOI: 10.3389/fmolb.2021.687319] [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: 03/29/2021] [Accepted: 11/03/2021] [Indexed: 12/24/2022] Open
Abstract
Background: Semaphorin 6b (SEMA6B) is a member of the semaphorin axon-guidance family and has been demonstrated to both induce and inhibit tumor progression. However, the role of SEMA6B in colorectal cancer (CRC) has remained unclear. This study sought to explore the promising prognostic biomarker for CRC and to understand the expression pattern, clinical significance, immune effects, and biological functions of SEMA6B. Methods: SEMA6B expression in CRC was evaluated via multiple gene and protein expression databases and we identified its prognostic value through The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Correlations between SEMA6B expression and components of the tumor immune microenvironment were analyzed by packages implemented in R, Tumor Immune Estimation Resource (TIMER), Gene Expression Profiling Interactive Analysis (GEPIA), and Tumor-Immune System Interactions database (TISIDB). RNA interference was performed to silence the expression of SEMA6B to explore its biological roles in the colon cancer cell lines HCT116 and LoVo. Results: The messenger RNA (mRNA) level of SEMA6B and the protein expression were higher in CRC tissues than adjacent normal tissues from multiple CRC datasets. High SEMA6B expression was significantly associated with dismal survival. Multivariate Cox regression analysis demonstrated that SEMA6B was an independent prognostic factor for progression-free survival (PFS). The nomogram showed a favorable predictive ability in PFS. Functional enrichment analysis and the Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE) algorithm revealed that the gene cluster associated with the high SEMA6B group were prominently involved in immune responses and inflammatory activities. Notably, SEMA6B expression was positively correlated with infiltrating levels of CD4+ T cells, macrophages, myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs), neutrophils, and dendritic cells. Moreover, SEMA6B expression displayed strong correlations with diverse marker sets of immunosuppressive cells in CRC. Integrative analysis revealed that immunosuppressive molecules and immune checkpoints were markedly upregulated in CRC samples with high SEMA6B expression. Furthermore, knockdown of SMEA6B in colon cancer cells significantly inhibited cell proliferation, migration, invasion and reduced the mRNA levels of immunosuppressive molecules. Conclusion: Our findings provide evidence that high SEMA6B expression correlated with adverse prognosis and the tumor immunosuppressive microenvironment in CRC patients. Therefore, SEMA6B may serve as a novel prognostic biomarker for CRC, which offers further insights into developing CRC-targeted immunotherapies.
Collapse
Affiliation(s)
- Tiegang Li
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Yan
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weiqi Wang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rixin Zhang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenqiang Gan
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Silin Lv
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zifan Zeng
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yufang Hou
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Min Yang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
65
|
Zhang J, Li S, Zhang X, Li C, Zhang J, Zhou W. LncRNA HLA-F-AS1 promotes colorectal cancer metastasis by inducing PFN1 in colorectal cancer-derived extracellular vesicles and mediating macrophage polarization. Cancer Gene Ther 2021; 28:1269-1284. [PMID: 33531647 DOI: 10.1038/s41417-020-00276-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 11/04/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) is a prevalent malignancy with high incidence and low 5-year survival. Long non-coding RNAs (lncRNAs), a kind of specific RNA transcript, are increasingly implicated in tumor growth, metastasis, invasion, and prognosis by regulating the tumor microenvironment in extracellular vesicles (EVs). This study aims at investigating the potential effect of lncRNA HLA-F-AS1 on CRC by affecting the profilin 1 (PFN1) expression pattern in the tumor EVs. The expression patterns of HLA-F-AS1 and miR-375 were determined by RT-qPCR in the CRC tissues and cells. CCK-8 and Transwell assays were conducted to detect the cell proliferation and migration, and invasion, respectively. Western blot analysis was performed to measure the expression pattern of the epithelial-mesenchymal transition (EMT) markers. Bioinformatics prediction website and dual-luciferase reporter assay were conducted to verify the interaction between HLA-F-AS1 and miR-375. The CRC-derived EVs were extracted with the expression pattern of PFN1 determined by ELISA, while its effect on the macrophage polarization was assessed by flow cytometry. The effect of PFN1-treated macrophages on CRC cell proliferation and migration was observed by subcutaneous tumorigenesis experiments in nude mice. The results indicated that the HLA-F-AS1 expression pattern was increased in the CRC tissues and cells, which promoted the migration, invasion, and EMT of CRC cells in vitro. Mechanistically, HLA-F-AS1 competitively bound to miR-375 and inversely regulated miR-375 expression pattern. Interestingly, PFN1 was identified as a direct target of miR-375, and positively modulated by HLA-F-AS1 by binding to miR-375. Overexpression of HLA-F-AS1 repressed miR-375 and promoted the PFN1 expression pattern in CRC cells and CRC-derived EVs, further promoting M2 polarization of macrophages. Furthermore, macrophages treated with PFN1 in CRC-derived EVs stimulated CRC cell proliferation and migration in vitro and in vivo. Collectively, these outcomes highlight that HLA-F-AS1 promotes the expression pattern of PFN1 in CRC-EVs by inhibiting miR-375, thereby polarizing macrophages toward M2 phenotype, and aggravating the tumorigenesis of CRC, eliciting that HLA-F-AS1 may serve as a viable and promising therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Colorectal and Anal Surgery, The First Hospital of Jilin University, Changchun, 130000, P.R. China
| | - Shiquan Li
- Department of Colorectal and Anal Surgery, The First Hospital of Jilin University, Changchun, 130000, P.R. China
| | - Xiaona Zhang
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, 130000, P.R. China
| | - Chao Li
- Department of Colorectal and Anal Surgery, The First Hospital of Jilin University, Changchun, 130000, P.R. China
| | - Jiantao Zhang
- Department of Colorectal and Anal Surgery, The First Hospital of Jilin University, Changchun, 130000, P.R. China.
| | - Wenli Zhou
- Department of Neonatology, The First Hospital of Jilin University, Changchun, 130000, P.R. China.
| |
Collapse
|
66
|
Lv Q, Pan X, Wang D, Rong Q, Ma B, Xie X, Zhang Y, Wang J, Hu L. Discovery of ( Z)-1-(3-((1 H-Pyrrol-2-yl)methylene)-2-oxoindolin-6-yl)-3-(isoxazol-3-yl)urea Derivatives as Novel and Orally Highly Effective CSF-1R Inhibitors for Potential Colorectal Cancer Immunotherapy. J Med Chem 2021; 64:17184-17208. [PMID: 34735158 DOI: 10.1021/acs.jmedchem.1c01184] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Inhibiting the polarization or survival of tumor-associated macrophages through blocking CSF-1/CSF-1R signal transduction has become a promising strategy for cancer immunotherapy. Herein, a series of (Z)-1-(3-((1H-pyrrol-2-yl)methylene)-2-oxoindolin-6-yl)-3-(isoxazol-3-yl)urea derivatives were designed, synthesized, and evaluated as novel and orally highly effective CSF-1R inhibitors for colorectal cancer immunotherapy. Among these derivatives, compound 21 was found to possess excellent CSF-1R inhibitory activity (IC50 = 2.1 nM) and potent antiproliferative activity against colorectal cancer cells. Compound 21 inhibited the progression of colorectal cancer by suppressing the migration of macrophages, reprograming M2-like macrophages to the M1 phenotype, and enhancing the antitumor immunity. More importantly, compound 21, as a single agent, showed significantly superior in vivo anticolorectal cancer efficacy over PLX3397, highlighting a promising candidate for the immunotherapy of colorectal cancer.
Collapse
Affiliation(s)
- Qi Lv
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Xiang Pan
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Dan Wang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Quanjin Rong
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Ben Ma
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Xiaolong Xie
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Yinan Zhang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Junwei Wang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Lihong Hu
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| |
Collapse
|
67
|
Wei X, Yao Y, Wang X, Sun J, Zhao W, Qiu L, Zhai W, Qi Y, Gao Y, Wu Y. Interleukin-36α inhibits colorectal cancer metastasis by enhancing the infiltration and activity of CD8 + T lymphocytes. Int Immunopharmacol 2021; 100:108152. [PMID: 34555640 DOI: 10.1016/j.intimp.2021.108152] [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: 06/30/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 02/07/2023]
Abstract
Colorectal cancer is one of the deadliest cancers, and the discovery of new diagnostic biomarkers and therapeutic targets is vital. Interleukin-36α (IL-36α) is a proinflammatory factor that can initiate the inflammatory response and promote the systemic T helper-1 (Th1) immune response. In this study, we investigated the immunological role of IL-36α in CRC. We found that IL-36α was downregulated in human CRC tissues. Patients with high IL-36α levels showed better survival and low IL-36α expression was significantly associated with greater tumor distal metastasis and TNM stage. We constructed two cell lines overexpressing IL-36α (CT26-IL-36α and HT29-IL-36α cells). In vitro assays revealed that IL-36α overexpression reduced the proliferation, migration, and invasion of CT26-IL-36α, and HT29-IL-36α cells. Using CT26-vector and CT26-IL-36α tumor mouse model and lung metastasis models, we found that IL-36α overexpression elicited a significant antitumor effect and inhibited lung metastasis in vivo. These inhibitory effects were associated with an increase in the number of CD3+CD8+ T lymphocytes within the tumor tissue as well as increased cytokine production in CD8+ T lymphocytes present in the tumor, spleen, and draining lymph nodes. Furthermore, we revealed that CT26-IL-36α cells enhanced the secretion of CXCL10 and CXCL11 from chemotactic CD8+ T lymphocytes, as compared with CT26-vector cells. Taken together, these results suggest that IL-36α is a promising therapeutic agent for targeting CRC by promoting the activation, proliferation, and tumor infiltration of T lymphocytes.
Collapse
Affiliation(s)
- Xiuyu Wei
- School of Life Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Yongjie Yao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Xiaoxi Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Jiaxin Sun
- School of Life Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Wenshan Zhao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450000, China; Henan Key Laboratory of Bioactive Macromolecules, Zhengzhou University, Zhengzhou 450001, China
| | - Lu Qiu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450000, China; Henan Key Laboratory of Bioactive Macromolecules, Zhengzhou University, Zhengzhou 450001, China
| | - Wenjie Zhai
- School of Life Sciences, Zhengzhou University, Zhengzhou 450000, China; Henan Key Laboratory of Bioactive Macromolecules, Zhengzhou University, Zhengzhou 450001, China
| | - Yuanming Qi
- School of Life Sciences, Zhengzhou University, Zhengzhou 450000, China; Henan Key Laboratory of Bioactive Macromolecules, Zhengzhou University, Zhengzhou 450001, China
| | - Yanfeng Gao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450000, China; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China; International Joint Laboratory for Protein and Peptide Drugs of Henan Province, Zhengzhou University, Zhengzhou 450001, China.
| | - Yahong Wu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450000, China; International Joint Laboratory for Protein and Peptide Drugs of Henan Province, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
68
|
Dhar R, Seethy A, Singh S, Pethusamy K, Srivastava T, Talukdar J, Rath GK, Karmakar S. Cancer immunotherapy: Recent advances and challenges. J Cancer Res Ther 2021; 17:834-844. [PMID: 34528529 DOI: 10.4103/jcrt.jcrt_1241_20] [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] [Indexed: 11/04/2022]
Abstract
Immunotherapy is a treatment that uses specific components of a person's immune system to fight diseases. This is usually done by stimulating or assisting one's immune system is attacking the offending agent - for instance, in the case of cancer - the target of immunotherapy will be cancer cells. Some types of immunotherapy are also called biologic therapy or biotherapy. One of the fundamental challenges that a living cell encounters are to accurately copy its genetic material to daughter cells during every single cell cycle. When this process goes haywire, genomic instability ensues, and genetic alterations ranging from nucleotide changes to chromosomal translocations and aneuploidy occur. Genomic instability arising out of DNA structural changes (indels, rearrangements, etc.,) can give rise to mutations predisposing to cancer. Cancer prevention refers to actions taken to mitigate the risk of getting cancer. The past decade has encountered an explosive rate of development of anticancer therapy ranging from standard chemotherapy to novel targeted small molecules that are nearly cancer specific, thereby reducing collateral damage. However, a new class of emerging therapy aims to train the body's defense system to fight against cancer. Termed as "cancer immunotherapy" is the new approach that has gained worldwide acceptance. It includes using antibodies that bind to and inhibit the function of proteins expressed by cancer cells or engineering and boosting the person's own T lymphocytes to target cancer. In this review, we summarized the recent advances and developments in cancer immunotherapy along with their shortcoming and challenges.
Collapse
Affiliation(s)
- Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Ashikh Seethy
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi; Department of Biochemistry, All India Institute of Medical Sciences, Guwahati, India
| | - Sunil Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Karthikeyan Pethusamy
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Tryambak Srivastava
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Joyeeta Talukdar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Goura Kishor Rath
- Department of Radiation Oncology, DRBRAIRCH, All India Institute of Medical Sciences, New Delhi; Department of Radiation Oncology, NCI, All India Institute of Medical Sciences, Jhajjar, Haryana, India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
69
|
Zheng Y, Zeng J, Xia H, Wang X, Chen H, Huang L, Zeng C. Upregulated lncRNA Cyclin-dependent kinase inhibitor 2B antisense RNA 1 induces the proliferation and migration of colorectal cancer by miR-378b/CAPRIN2 axis. Bioengineered 2021; 12:5476-5490. [PMID: 34511033 PMCID: PMC8806871 DOI: 10.1080/21655979.2021.1961656] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
LncRNA Cyclin‐dependent kinase inhibitor 2B antisense RNA 1 (CDKN2B-AS1) plays a role in the progression of multiple cancers like cholangiocarcinoma, osteosarcoma and several gastrointestinal tumors. Few studies have linked its function and mechanism to the development of colorectal cancer (CRC). The expression of CDKN2B-AS1, microRNA (miR)-378b, and cytoplasmic activation/proliferation-associated protein 2 (CAPRIN2) was analyzed in CRC patients and cell lines. The proliferation and migration of CRC cells were evaluated after gain and loss-of function mutations. Interactions between CDKN2B-AS1 and miR-378b, miR-378b and CAPRIN2 were validated by luciferase reporter, RNA pull-down and RNA immunoprecipitation assays. The role of CDKN2B-AS1 was further confirmed in a xenograft mouse model. We found that the expression of CDKN2B-AS1 and CAPRIN2 was upregulated in CRC and they were linked to the poor differentiation and distant metastasis in CRC patients. CDKN2B-AS1 knockdown attenuated while CDKN2B-AS1 overexpression promoted CRC cell proliferation and migration. Notably, the results of Starbase 2.0 database analysis and in vitro experiments demonstrated that CDKN2B-AS1 could interact with miR-378b and regulate its expression. Furthermore, CAPRIN2 acted as a downstream target of CDKN2B-AS1/miR-378b that involved in modulating β-catenin expression in CRC cells. Upregulation of CDKN2B-AS1 contributed to CRC progression via regulating CAPRIN2 expression by binding to miR-378b. Downregulation of CDKN2B-AS1 suppressed tumor growth and Ki-67 staining in vivo that was related to the miR-378b/CAPRIN2 pathway. This study indicated that lncRNA CDKN2B-AS1 promoted the development of CRC through the miR-378b/CAPRIN2/β-catenin axis. CDKN2B-AS1 might serve as a potential and useful target in CRC diagnosis and treatment.
Collapse
Affiliation(s)
- Yu Zheng
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Provincial Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Jintao Zeng
- Department of Clinical Medicine, School of Basic Medicine, Chengde Medical College, Chengde, China
| | - Haoyun Xia
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Provincial Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Xiangyu Wang
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Provincial Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Hongyuan Chen
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Provincial Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Liangxiang Huang
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Provincial Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Changqing Zeng
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Provincial Clinical Medical College of Fujian Medical University, Fuzhou, China
| |
Collapse
|
70
|
Zhuang QS, Sun XB, Chong QY, Banerjee A, Zhang M, Wu ZS, Zhu T, Pandey V, Lobie PE. ARTEMIN Promotes Oncogenicity and Resistance to 5-Fluorouracil in Colorectal Carcinoma by p44/42 MAPK Dependent Expression of CDH2. Front Oncol 2021; 11:712348. [PMID: 34422665 PMCID: PMC8377398 DOI: 10.3389/fonc.2021.712348] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/07/2021] [Indexed: 12/12/2022] Open
Abstract
ARTEMIN (ARTN), one of the glial-cell derived neurotrophic factor family of ligands, has been reported to be associated with a number of human malignancies. In this study, the enhanced expression of ARTN in colorectal carcinoma (CRC) was observed; the expression of ARTN positively correlated with lymph node metastases and advanced tumor stages and predicted poor prognosis. Forced expression of ARTN in CRC cells enhanced oncogenic behavior, mesenchymal phenotype, stem cell-like properties and tumor growth and metastasis in a xenograft model. These functions were conversely inhibited by depletion of endogenous ARTN. Forced expression of ARTN reduced the sensitivity of CRC cells to 5-FU treatment; and 5-FU resistant CRC cells harbored enhanced expression of ARTN. The oncogenic functions of ARTN were demonstrated to be mediated by p44/42 MAP kinase dependent expression of CDH2 (CADHERIN 2, also known as N-CADHERIN). Inhibition of p44/42 MAP kinase activity or siRNA mediated depletion of endogenous CDH2 reduced the enhanced oncogenicity and chemoresistance consequent to forced expression of ARTN induced cell functions; and forced expression of CDH2 rescued the reduced mesenchymal properties and resistance to 5-FU after ARTN depletion. In conclusion, ARTN may be of prognostic and theranostic utility in CRC.
Collapse
Affiliation(s)
- Qiu-Shi Zhuang
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore, Singapore.,Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China.,Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Xin-Bao Sun
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Qing-Yun Chong
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore, Singapore
| | - Arindam Banerjee
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore, Singapore.,Department of Chemical Engineering, Indian Institute of Technology, Kharagpur, India
| | - Min Zhang
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Zheng-Sheng Wu
- Department of Pathology, Anhui Medical University, Hefei, China
| | - Tao Zhu
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Vijay Pandey
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China.,Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Peter E Lobie
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore, Singapore.,Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China.,Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China.,Shenzhen Bay Laboratory, Shenzhen, China
| |
Collapse
|
71
|
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
|
72
|
The State of Immunotherapy in Hepatobiliary Cancers. Cells 2021; 10:cells10082096. [PMID: 34440865 PMCID: PMC8393650 DOI: 10.3390/cells10082096] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 08/02/2021] [Accepted: 08/13/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatobiliary cancers, including hepatocellular carcinoma (HCC), cholangiocarcinoma (CCA), and gallbladder carcinoma (GBC), are lethal cancers with limited therapeutic options. Curative-intent treatment typically involves surgery, yet recurrence is common and many patients present with advanced disease not amenable to an operation. Immunotherapy represents a promising approach to improve outcomes, but the immunosuppressive tumor microenvironment of the liver characteristic of hepatobiliary cancers has hampered the development and implementation of this therapeutic approach. Current immunotherapies under investigation include immune checkpoint inhibitors (ICI), the adoptive transfer of immune cells, bispecific antibodies, vaccines, and oncolytic viruses. Programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) are two ICIs that have demonstrated utility in HCC, and newer immune checkpoint targets are being tested in clinical trials. In advanced CCA and GBC, PD-1 ICIs have resulted in antitumor responses, but only in a minority of select patients. Other ICIs are being investigated for patients with CCA and GBC. Adoptive transfer may hold promise, with reports of complete durable regression in metastatic CCA, yet this therapeutic approach may not be generalizable. Alternative approaches have been developed and promising results have been observed, but clinical trials are needed to validate their utility. While the treatment of hepatobiliary cancers involves unique challenges that these cancers present, the progress seen with ICIs and adoptive transfer has solidified immunotherapy as an important approach in these challenging patients with few other effective treatment options.
Collapse
|
73
|
Galbraith NJ, Wood C, Steele CW. Targeting Metastatic Colorectal Cancer with Immune Oncological Therapies. Cancers (Basel) 2021; 13:3566. [PMID: 34298779 PMCID: PMC8307556 DOI: 10.3390/cancers13143566] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/08/2021] [Accepted: 07/11/2021] [Indexed: 02/07/2023] Open
Abstract
Metastatic colorectal cancer carries poor prognosis, and current therapeutic regimes convey limited improvements in survival and high rates of detrimental side effects in patients that may not stand to benefit. Immunotherapy has revolutionised cancer treatment by restoring antitumoural mechanisms. However, the efficacy in metastatic colorectal cancer, is limited. A literature search was performed using Pubmed (Medline), Web of Knowledge, and Embase. Search terms included combinations of immunotherapy and metastatic colorectal cancer, primarily focusing on clinical trials in humans. Analysis of these studies included status of MMR/MSS, presence of combination strategies, and disease control rate and median overall survival. Evidence shows that immune checkpoint inhibitors, such as anti-PD1 and anti-PD-L1, show efficacy in less than 10% of patients with microsatellite stable, MMR proficient colorectal cancer. In the small subset of patients with microsatellite unstable, MMR deficient cancers, response rates were 40-50%. Combination strategies with immunotherapy are under investigation but have not yet restored antitumoural mechanisms to permit durable disease regression. Immunotherapy provides the potential to offer additional strategies to established chemotherapeutic regimes in metastatic colorectal cancer. Further research needs to establish which adjuncts to immune checkpoint inhibition can unpick resistance, and better predict which patients are likely to respond to individualised therapies to not just improve response rates but to temper unwarranted side effects.
Collapse
Affiliation(s)
- Norman J. Galbraith
- Academic Department of Surgery, University of Glasgow, Level 2 New Lister Building, Glasgow Royal Infirmary, 10-16 Alexandra Parade, Glasgow G31 2ER, UK; (C.W.); (C.W.S.)
| | - Colin Wood
- Academic Department of Surgery, University of Glasgow, Level 2 New Lister Building, Glasgow Royal Infirmary, 10-16 Alexandra Parade, Glasgow G31 2ER, UK; (C.W.); (C.W.S.)
| | - Colin W. Steele
- Academic Department of Surgery, University of Glasgow, Level 2 New Lister Building, Glasgow Royal Infirmary, 10-16 Alexandra Parade, Glasgow G31 2ER, UK; (C.W.); (C.W.S.)
- Institute of Cancer Sciences, Beatson Institute, Garscube Campus, Switchback Road, Bearsden G61 1BD, UK
| |
Collapse
|
74
|
The Clinical Value of the Combined Detection of Enhanced CT, MRI, CEA, and CA199 in the Diagnosis of Rectal Cancer. JOURNAL OF ONCOLOGY 2021; 2021:8585371. [PMID: 34335762 PMCID: PMC8292063 DOI: 10.1155/2021/8585371] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/05/2021] [Indexed: 12/19/2022]
Abstract
Background To explore the clinical value of enhanced computed tomography (enhanced CT), magnetic resonance imaging (MRI), carcinoembryonic antigen (CEA), and cancer antigen 199 (CA199) in the diagnosis of rectal cancer (RC). Methods A total of 156 patients with RC confirmed by postoperative pathology admitted to the Affiliated Yantai Yuhuangding Hospital of Qingdao University from March 2018 to November 2020 were included in the malignant group, and 52 patients with chronic proctitis in the benign control group. All patients underwent preoperative enhanced CT, MRI scans, and serum CEA and CA199 tests. The accuracy, sensitivity, and specificity of single and combined enhanced CT, MRI, CEA, and CA199 tests for the clinical staging of RC were calculated. Results The postoperative pathological diagnosis showed that 35 cases of 156 RC patients were at T1 stage, 29 cases were at T2 stage, 24 cases were at T3 stage, 11 cases were at T4 stage, 23 cases were at N0 stage, 21 cases were at N1 stage, 8 cases were at N2 stage, 3 cases were at M0 stage, and 2 cases were at M1 stage. The positive rate of MRI in the diagnosis of RC was higher than that of enhanced CT. Serum CEA and CA199 levels in the malignant group were significantly increased compared with the benign group. The sensitivity, specificity, and accuracy of the combined detection were significantly higher than those of the single detection. Conclusion Compared with enhanced CT, MRI has a higher detection rate of T and N stage in patients with RC. Combined enhanced CT, MRI, CEA, and CA199 can provide more accurate diagnosis and preoperative staging of RC patients.
Collapse
|
75
|
Shao S, Ju M, Lei J, Lu X, Li H, Wang D, Xia C. Egr‑1 inhibits colon cancer cell proliferation, migration and invasion via regulating CDKL1 at the transcriptional level. Oncol Rep 2021; 46:169. [PMID: 34165179 DOI: 10.3892/or.2021.8120] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/21/2021] [Indexed: 11/05/2022] Open
Abstract
Colon cancer is one of the most common malignant tumors worldwide, and the molecular mechanisms involved in the oncogenesis and progression of colon cancer remain unclear. Early growth response 1 (Egr‑1) is a transcription factor that is closely associated with several tumor processes; however, its role in colon cancer is unknown. The present study aimed to explore the function and mechanism of transcription factor Egr‑1 in colon cancer progression. The association between Egr‑1 expression and the survival of patients with colon cancer was analyzed. Transwell assay was used to measure the migration and invasion of colon cancer cells. Cell Counting Kit‑8 assay was used to evaluate the cell proliferative ability. Reverse transcription‑quantitative PCR and western blot assays were used to identify whether Egr‑1 could regulate cyclin‑dependent kinase‑like 1 (CDKL1). Luciferase and chromatin immunoprecipitation assays were used to detect the mechanism by which Egr‑1 regulated CDKL1. Based on The Cancer Genome Atlas database, it was found that low Egr‑1 expression was associated with a poor prognosis in patients with colon cancer. Furthermore, overexpression of Egr‑1 inhibited colon cancer cell proliferation, migration, and invasion, whereas knockdown of Egr‑1 increased colon cancer cell proliferation, migration and invasion. Additionally, overexpression of Egr‑1‑induced cell proliferation, migration and invasion were reversed by overexpression of CDKL1. Furthermore, it was demonstrated that Egr‑1 regulated CDKL1 expression at the transcriptional level. The present study illustrated the mechanism of Egr‑1 regulating CDKL1, by which Egr‑1 affected colon cancer cell proliferation, migration and invasion. The current findings suggested that Egr‑1/CDKL1 may be a new promising target for the treatment of colon cancer.
Collapse
Affiliation(s)
- Shanshan Shao
- Department of Oncology, The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People's Hospital), Taizhou, Jiangsu 225300, P.R. China
| | - Man Ju
- Department of Anus and Intestine Surgery, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Jiayun Lei
- Department of Oncology, Liaocheng Dongchangfu People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Xiangqian Lu
- Department of Radiotherapy, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Hongzhi Li
- Department of Oncology, The Hospital Affiliated to Medical School of Yangzhou University (Taizhou People's Hospital), Taizhou, Jiangsu 225300, P.R. China
| | - Darui Wang
- Department of Clinical Laboratory, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Chaofeng Xia
- Department of Anus and Intestine Surgery, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| |
Collapse
|
76
|
Stamova S, Ott-Rötzer B, Smetak H, Schäffler K, Eder R, Fink I, Hoffmann P, Reichert TE, Beckhove P, Spanier G. Characterization and ex vivo expansion of rare in situ cytokine secreting T cell populations from tumor tissue and blood of oral squamous cell carcinoma patients. J Immunol Methods 2021; 496:113086. [PMID: 34146580 DOI: 10.1016/j.jim.2021.113086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 06/14/2021] [Accepted: 06/14/2021] [Indexed: 12/18/2022]
Abstract
Rare subpopulations of tumor antigen-reactive memory T cells, which actively secrete type-1 effector cytokines, particularly TNF-α in situ, possess anti-tumor activity and prognostic relevance. These cells are relevant for cancer immunotherapy; however, their low frequencies make them difficult to study and novel protocols for their culture and expansion ex vivo are needed. Here, we studied the presence of T cells secreting type-1 cytokines (Cy+T cells) in the blood and tumors of 24 patients with oral squamous cell carcinomas (OSCC) and explored possibilities for their isolation and expansion. More than 90% of OSCC patients contained enriched numbers Cy+T cells in the blood and tumors compared to healthy donors in which these were hardly detectable. The majority of TNF-α+T cells were CD4+ T helper cells while IFN-γ+TIL were predominantly CD8+. Cy+T helper cells in the blood were early-differentiated memory T cells while Cy+TIL and Cy+CD8+T cells showed advanced-differentiated memory T cell phenotypes. We explored different conditions for their in vitro culture and found that Cy+T cells can be efficiently expanded in vitro to similar levels as Cy-T cells and after expansion maintained their TNF-α secreting capacity. However, for optimal expansion they required specific culture conditions to support the maintenance of stem-like and central memory T cell phenotype. In conclusion, we show that Cy+T cells are enriched in OSCC patients and report a novel cell culture protocol optimized to specifically expand and functionally maintain these cells for further functional characterization or for their exploitation in immunotherapy of OSCC.
Collapse
Affiliation(s)
- Slava Stamova
- Regensburg Center for Interventional Immunology (RCI), University Hospital Regensburg, Regensburg, Germany
| | - Birgitta Ott-Rötzer
- Regensburg Center for Interventional Immunology (RCI), University Hospital Regensburg, Regensburg, Germany
| | - Heiko Smetak
- Regensburg Center for Interventional Immunology (RCI), University Hospital Regensburg, Regensburg, Germany
| | - Katharina Schäffler
- Regensburg Center for Interventional Immunology (RCI), University Hospital Regensburg, Regensburg, Germany
| | - Rüdiger Eder
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Irina Fink
- Regensburg Center for Interventional Immunology (RCI), University Hospital Regensburg, Regensburg, Germany
| | - Petra Hoffmann
- Regensburg Center for Interventional Immunology (RCI), University Hospital Regensburg, Regensburg, Germany; Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Torsten E Reichert
- Department of Cranio-Maxillofacial Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Philipp Beckhove
- Regensburg Center for Interventional Immunology (RCI), University Hospital Regensburg, Regensburg, Germany; Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany.
| | - Gerrit Spanier
- Department of Cranio-Maxillofacial Surgery, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
77
|
Mughal MJ, Kwok HF. Multidimensional role of bacteria in cancer: Mechanisms insight, diagnostic, preventive and therapeutic potential. Semin Cancer Biol 2021; 86:1026-1044. [PMID: 34119644 DOI: 10.1016/j.semcancer.2021.06.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/28/2021] [Accepted: 06/08/2021] [Indexed: 02/08/2023]
Abstract
The active role of bacteria in oncogenesis has long been a topic of debate. Although, it was speculated to be a transmissible cause of cancer as early as the 16th-century, yet the idea about the direct involvement of bacteria in cancer development has only been explored in recent decades. More recently, several studies have uncovered the mechanisms behind the carcinogenic potential of bacteria which are inflammation, immune evasion, pro-carcinogenic metabolite production, DNA damage and genomic instability. On the other side, the recent development on the understanding of tumor microenvironment and technological advancements has turned this enemy into an ally. Studies using bacteria for cancer treatment and detection have shown noticeable effects. Therapeutic abilities of bioengineered live bacteria such as high specificity, selective cytotoxicity to cancer cells, responsiveness to external signals and control after ingestion have helped to overcome the challenges faced by conventional cancer therapies and highlighted the bacterial based therapy as an ideal approach for cancer treatment. In this review, we have made an effort to compile substantial evidence to support the multidimensional role of bacteria in cancer. We have discussed the multifaceted role of bacteria in cancer by highlighting the wide impact of bacteria on different cancer types, their mechanisms of actions in inducing carcinogenicity, followed by the diagnostic and therapeutic potential of bacteria in cancers. Moreover, we have also highlighted the existing gaps in the knowledge of the association between bacteria and cancer as well as the limitation and advantage of bacteria-based therapies in cancer. A better understanding of these multidimensional roles of bacteria in cancer can open up the new doorways to develop early detection strategies, prevent cancer, and develop therapeutic tactics to cure this devastating disease.
Collapse
Affiliation(s)
- Muhammad Jameel Mughal
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau
| | - Hang Fai Kwok
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau; MOE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau.
| |
Collapse
|
78
|
Tang S, Liu Q, Xu M. LINC00857 promotes cell proliferation and migration in colorectal cancer by interacting with YTHDC1 and stabilizing SLC7A5. Oncol Lett 2021; 22:578. [PMID: 34122629 PMCID: PMC8190780 DOI: 10.3892/ol.2021.12839] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/12/2021] [Indexed: 12/15/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most lethal malignances in humans. Hence, it is of great significance to identify regulatory molecules in CRC progression. Accumulating evidence has demonstrated that long non-coding RNAs (lncRNAs) are involved in cancer malignancy. It has been reported that long intergenic non-protein coding RNA 857 (LINC00857) acts as a vital oncogene in many types of cancer by promoting cell proliferation and migration. However, the role of LINC00857 in CRC remains unclear. In the present study, LINC00857 was upregulated in CRC tissue samples and cells. Next, in vitro loss-of-function experiments demonstrated that LINC00857 knockdown suppressed CRC cell viability, proliferation and migration, as well as epithelial-mesenchymal transition and increased cell apoptosis. Mechanistically, LINC00857 abundantly interacted with the RNA-binding protein YTH domain containing 1 (YTHDC1). YTHDC1 ultimately combined with solute carrier family 7 member 5 (SLC7A5) and increased SLC7A5 mRNA stability. Finally, a series of rescue experiments indicated that LINC00857 promoted the proliferation and migration of CRC cells by regulating mRNA stability. Thus, the present findings illustrated that LINC00857 functions as an oncogene in CRC cells via the YTHDC1/SLC7A5 axis.
Collapse
Affiliation(s)
- Shu Tang
- Department of Internal Medicine-Oncology, Chenzhou No. 1 People's Hospital, Chenzhou, Hunan 423000, P.R. China
| | - Qi Liu
- Hospital Medical Department, Chenzhou No. 1 People's Hospital, Chenzhou, Hunan 423000, P.R. China
| | - Ming Xu
- Department of Gastrointestinal Surgery, Chenzhou No. 1 People's Hospital, Chenzhou, Hunan 423000, P.R. China
| |
Collapse
|
79
|
Zheng W, Wu F, Fu K, Sun G, Sun G, Li X, Jiang W, Cao H, Wang H, Tang W. Emerging Mechanisms and Treatment Progress on Liver Metastasis of Colorectal Cancer. Onco Targets Ther 2021; 14:3013-3036. [PMID: 33986602 PMCID: PMC8110277 DOI: 10.2147/ott.s301371] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 03/24/2021] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer is currently the third largest malignant tumor in the world, with high new cases and high mortality. Metastasis is one of the most common causes of death of colorectal cancer, of which liver metastasis is the most fatal. Since the beginning of the Human Genome Project in 2001, people have gradually recognized the 3 billion base pairs that make up the human genome, of which only about 1.5% of the nucleic acid sequences are used for protein coding, including proto-oncogenes and tumor suppressor genes. A large number of differences in the expression of proto-oncogenes and tumor suppressor genes have also been found in the study of colorectal cancer, which proves that they are also actively involved in the progression of colorectal cancer and promote the occurrence of liver metastasis. Except for 1.5% of the coding sequence, the rest of the nucleic acid sequence does not encode any protein, which is called non-coding RNA. With the deepening of research, genome sequences without protein coding potential that were originally considered “junk sequences” may have important biological functions. Many years of studies have found that a large number of abnormal expression of ncRNA in colorectal cancer liver metastasis, indicating that ncRNA plays an important role in it. To explore the role and mechanism of these coding sequences and non-coding RNA in liver metastasis of colorectal cancer is very important for the early diagnosis and treatment of liver metastasis of colorectal cancer. This article reviews the coding genes and ncRNA that have been found in the study of liver metastasis of colorectal cancer in recent years, as well as the mechanisms that have been identified or are still under study, as well as the clinical treatment of liver metastasis of colorectal cancer.
Collapse
Affiliation(s)
- Wubin Zheng
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Fan Wu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Kai Fu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Guangshun Sun
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Guoqiang Sun
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Xiao Li
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Wei Jiang
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Hongyong Cao
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Hanjin Wang
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Weiwei Tang
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Transplantation, Chinese Academy of Medical Sciences, Nanjing, People's Republic of China
| |
Collapse
|
80
|
Feng Y, Jin H, Guo K, Wasan HS, Ruan S, Chen C. Causes of Death After Colorectal Cancer Diagnosis: A Population-Based Study. Front Oncol 2021; 11:647179. [PMID: 33859947 PMCID: PMC8042257 DOI: 10.3389/fonc.2021.647179] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/11/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Non-cancer causes of death in patients with colorectal cancer (CRC) have not received much attention until now. The purpose of the current study is to investigate the non-cancer causes of death in patients with CRC at different periods of latency. Methods: Eligible patients with CRC were included from the Surveillance, Epidemiology, and End Results (SEER) database, and standardized mortality ratios (SMRs) were calculated using the SEER*Stat software 8.3.8. Results: A total of 475,771 patients with CRC were included, of whom 230,841 patients died during the follow-up period. Within 5 years, CRC was the leading cause of death. Over time, non-cancer causes of death account for an increasing proportion. When followed up for more than 10 years, non-cancer deaths accounted for 71.9% of all deaths worldwide. Cardiovascular diseases were the most common causes of non-cancer deaths, accounting for 15.4% of the total mortality. Patients had a significantly higher risk of death from septicemia within the first year after diagnosis compared with the general population (SMR, 3.39; 95% CI, 3.11–3.69). Within 5–10 years after CRC diagnosis, patients had a significantly higher risk of death from diabetes mellitus (SMR, 1.27; 95% CI, 1.19–1.36). During the course of more than 10 years, patients with CRC had a significantly higher risk of death from atherosclerosis (SMR 1.47; 95% CI, 1.11–1.9). Conclusions: Although CRC has always been the leading cause of death in patients with CRC, non-cancer causes of death should not be ignored. For patients with cancer, we should not only focus on anti-tumor therapies but also pay attention to the occurrence of other risks to prevent and manage them in advance.
Collapse
Affiliation(s)
- Yuqian Feng
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Huimin Jin
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Kaibo Guo
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Harpreet S Wasan
- Department of Cancer Medicine, Hammersmith Hospital, Imperial College Healthcare National Health Service Trust, London, United Kingdom
| | - Shanming Ruan
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Cihui Chen
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
81
|
Han W, Kong D, Lu Q, Zhang W, Fan Z. Aloperine Inhibits Proliferation and Promotes Apoptosis in Colorectal Cancer Cells by Regulating the circNSUN2/miR-296-5p/STAT3 Pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:857-870. [PMID: 33664565 PMCID: PMC7924259 DOI: 10.2147/dddt.s288473] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/16/2021] [Indexed: 12/14/2022]
Abstract
Background Aloperine can regulate miR-296-5p/Signal Transducer and Activator of Transcription 3 (STAT3) pathway to inhibit the malignant development of colorectal cancer (CRC), but the regulatory mechanism is unclear. This study explored the upstream mechanism of Aloperine in reducing CRC damage from the perspective of the circRNA-miRNA-mRNA regulatory network. Methods After treatment with gradient concentrations of Aloperine (0.1 mmol/L, 0.2 mmol/L, 0.4 mmol/L, 0.8 mmol/L and 1 mmol/L) for 24 hours, changes in CRC cell proliferation and apoptosis were detected by functional experiments. Data of the differential expression of miR-296-5p in CRC patients and healthy people were obtained from Starbase. The effects of Aloperine on 12 differentially expressed circRNAs were detected. The binding of miR-296-5p with NOP2/Sun RNA methyltransferase 2 (circNSUN2) and STAT3 was predicted by TargetScan and confirmed through dual-luciferase experiments. The expressions of circNSUN2, miR-296-5p and STAT3 as well as apoptosis-related genes in CRC cells were detected by qRT-PCR and Western blot as needed. Rescue experiments were conducted to test the regulatory effects of circNSUN2, miR-296-5p and STAT3 on CRC cells. Results Aloperine at a concentration gradient inhibited proliferation and promoted apoptosis in CRC cells. The abnormally low expression of miR-296-5p in CRC could be upregulated by Aloperine. Among the differentially expressed circRNAs in CRC, only circNSUN2 not only targets miR-296-5p, but also can be regulated by Aloperine. The up-regulation of circNSUN2 offset the inhibitory effect of Aloperine on cancer cells. The rescue experiments finally confirmed the regulation of circNSUN2/miR-296-5p/STAT3 axis in CRC cells. Conclusion By regulating the circNSUN2/miR-296-5p/STAT3 pathway, Aloperine prevents the malignant development of CRC cells.
Collapse
Affiliation(s)
- Wei Han
- Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,General Surgery Department, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Desong Kong
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Qin Lu
- Proctology Department, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Wei Zhang
- Anesthesiology Department, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Zhimin Fan
- Proctology Department, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| |
Collapse
|
82
|
TUFT1 Facilitates Metastasis, Stemness, and Vincristine Resistance in Colorectal Cancer via Activation of PI3K/AKT Pathway. Biochem Genet 2021; 59:1018-1032. [PMID: 33634374 DOI: 10.1007/s10528-021-10051-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/10/2021] [Indexed: 12/22/2022]
Abstract
Since the incidence and mortality of colorectal cancer (CRC) are increasing in recent years, the research on the pathogenesis of colorectal cancer has attracted more and more attention. Here, our results confirmed that the mRNA expression level and proteins accumulation of TUFT1 were significantly increased in CRC tissues from late-stage CRC patients (III + IV) (p < 0.001), indicated by qPCR and IHC assay. The TUFT1 expression was positively correlated with tumor stage by analyzing 126 specimens from CRC patients. Next, we found that up-regulation of TUFT1 enhanced the migration and invasion of LoVo cells, whereas the down-regulation of TUFT1 observably weakened the migration and invasion of SW837 cells, indicating that TUFT1 promotes the metastasis of CRC cells. In addition, TUFT1 overexpression increased the number of mammary spheres and vincristine resistance of LoVo cells by sphere formation assay and measuring the IC50 value, suggesting the TUFT1 promotes stemness and the vincristine resistance of CRC cells. Finally, we found that TUFT1 overexpression increased p-AKT in LoVo cells, while down-regulation of TUFT1 decreased the p-AKT levels in SW837 cells. Therefore, we determined that the function of TUFT1 in CRC depends on PI3K/AKT pathway. Taken together, these data demonstrated that TUFI1 facilitates metastasis, stemness, and vincristine resistance of colorectal cancer cells via activation of PI3K/AKT pathway, which might act as a promising therapeutic target for CRC.
Collapse
|
83
|
Xiong D, Wang D, Chen Y. Role of the long non-coding RNA LINC00052 in tumors. Oncol Lett 2021; 21:316. [PMID: 33692848 PMCID: PMC7933760 DOI: 10.3892/ol.2021.12577] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
Long intergenic non-protein coding RNA 52 (LINC00052) is a non-coding RNA with >200 nucleotides in length, which exerts important effects on several physiological and pathological processes of the human body. Recent studies have demonstrated that LINC00052 plays key roles in the tumorigenesis, progression and metastasis of multiple types of human cancer, including hepatocellular carcinoma, breast cancer, colorectal cancer, cervical carcinoma and gastric cancer. However, the associations between LINC00052 and these tumors remain unclear. The present review summarizes the biological functions of LINC00052 during the pathogenic process of certain tumors, and discusses its potential therapeutic targets.
Collapse
Affiliation(s)
- Dongmei Xiong
- Early Childhood Health Research Innovation Team, Nursing School of Chongqing Medical and Pharmaceutical College, Chongqing 401331, P.R. China
| | - Dan Wang
- Clinical Laboratory, The People's Hospital of Rongchang, Chongqing 402460, P.R. China
| | - Yanmeng Chen
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
84
|
Lisby AN, Flickinger JC, Bashir B, Weindorfer M, Shelukar S, Crutcher M, Snook AE, Waldman SA. GUCY2C as a biomarker to target precision therapies for patients with colorectal cancer. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2021; 6:117-129. [PMID: 34027103 DOI: 10.1080/23808993.2021.1876518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction Colorectal cancer (CRC) is one of the most-deadly malignancies worldwide. Current therapeutic regimens for CRC patients are relatively generic, based primarily on disease type and stage, with little variation. As the field of molecular oncology advances, so too must therapeutic management of CRC. Understanding molecular heterogeneity has led to a new-found promotion for precision therapy in CRC; underlining the diversity of molecularly targeted therapies based on individual tumor characteristics. Areas covered We review current approaches for the treatment of CRC and discuss the potential of precision therapy in advanced CRC. We highlight the utility of the intestinal protein guanylyl cyclase C (GUCY2C), as a multi-purpose biomarker and unique therapeutic target in CRC. Here, we summarize current GUCY2C-targeted approaches for treatment of CRC. Expert opinion The GUCY2C biomarker has multi-faceted utility in medicine. Developmental investment of GUCY2C as a diagnostic and therapeutic biomarker offers a variety of options taking the molecular characteristics of cancer into account. From GUCY2C-targeted therapies, namely cancer vaccines, CAR-T cells, and monoclonal antibodies, to GUCY2C agonists for chemoprevention in those who are at high risk for developing colorectal cancer, the utility of this protein provides many avenues for exploration with significance in the field of precision medicine.
Collapse
Affiliation(s)
- Amanda N Lisby
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - John C Flickinger
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Babar Bashir
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Megan Weindorfer
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Sanjna Shelukar
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Madison Crutcher
- Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Adam E Snook
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Scott A Waldman
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, United States
| |
Collapse
|
85
|
Pavitra E, Dariya B, Srivani G, Kang SM, Alam A, Sudhir PR, Kamal MA, Raju GSR, Han YK, Lakkakula BVKS, Nagaraju GP, Huh YS. Engineered nanoparticles for imaging and drug delivery in colorectal cancer. Semin Cancer Biol 2021; 69:293-306. [PMID: 31260733 DOI: 10.1016/j.semcancer.2019.06.017] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 06/18/2019] [Accepted: 06/27/2019] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) is one of the deadliest diseases worldwide due to a lack of early detection methods and appropriate drug delivery strategies. Conventional imaging techniques cannot accurately distinguish benign from malignant tissue, leading to frequent misdiagnosis or diagnosis at late stages of the disease. Novel screening tools with improved accuracy and diagnostic precision are thus required to reduce the mortality burden of this malignancy. Additionally, current therapeutic strategies, including radio- and chemotherapies carry adverse side effects and are limited by the development of drug resistance. Recent advances in nanotechnology have rendered it an attractive approach for designing novel clinical solutions for CRC. Nanoparticle-based formulations could assist early tumor detection and help to overcome the limitations of conventional therapies including poor aqueous solubility, nonspecific biodistribution and limited bioavailability. In this review, we shed light on various types of nanoparticles used for diagnosis and drug delivery in CRC. In addition, we will explore how these nanoparticles can improve diagnostic accuracy and promote selective drug targeting to tumor sites with increased efficiency and reduced cytotoxicity against healthy colon tissue.
Collapse
Affiliation(s)
- Eluri Pavitra
- Department of Biological Engineering, Biohybrid Systems Research Center (BSRC) Inha University, Incheon, 22212, Republic of Korea.
| | - Begum Dariya
- Department of Bioscience and Biotechnology, Banasthali University, Vanasthali, Rajasthan, 304022, India
| | - Gowru Srivani
- Department of Bioscience and Biotechnology, Banasthali University, Vanasthali, Rajasthan, 304022, India
| | - Sung-Min Kang
- Department of Biological Engineering, Biohybrid Systems Research Center (BSRC) Inha University, Incheon, 22212, Republic of Korea
| | - Afroz Alam
- Department of Bioscience and Biotechnology, Banasthali University, Vanasthali, Rajasthan, 304022, India
| | - Putty-Reddy Sudhir
- The Center for Translational Biomedical Research, UNCG, Kannapolis, NC-28081, USA
| | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Saudi Arabia; Enzymoics, 7 Peterlee Place, Hebersham, NSW, 2770, Australia; Novel Global Community Educational Foundation, Australia
| | - Ganji Seeta Rama Raju
- Department of Energy and Materials Engineering, Dongguk University-Seoul, Seoul, 04620, Republic of Korea
| | - Young-Kyu Han
- Department of Energy and Materials Engineering, Dongguk University-Seoul, Seoul, 04620, Republic of Korea
| | | | - Ganji Purnachandra Nagaraju
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA-30322, USA
| | - Yun Suk Huh
- Department of Biological Engineering, Biohybrid Systems Research Center (BSRC) Inha University, Incheon, 22212, Republic of Korea.
| |
Collapse
|
86
|
Gao W, Chen Y, Yang J, Zhuo C, Huang S, Zhang H, Shi Y. Clinical Perspectives on Liquid Biopsy in Metastatic Colorectal Cancer. Front Genet 2021; 12:634642. [PMID: 33584829 PMCID: PMC7876389 DOI: 10.3389/fgene.2021.634642] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 01/04/2021] [Indexed: 02/05/2023] Open
Abstract
Liquid biopsy, which generally refers to the analysis of biological components such as circulating nuclear acids and circulating tumor cells in body fluids, particularly in peripheral blood, has shown good capacity to overcome several limitations faced by conventional tissue biopsies. Emerging evidence in recent decades has confirmed the promising role of liquid biopsy in the clinical management of various cancers, including colorectal cancer, which is one of the most prevalent cancers and the second leading cause of cancer-related deaths worldwide. Despite the challenges and poor clinical outcomes, patients with metastatic colorectal cancer can expect potential clinical benefits with liquid biopsy. Therefore, in this review, we focus on the clinical prospects of liquid biopsy in metastatic colorectal cancer, specifically with regard to the recently discovered various biomarkers identified on liquid biopsy. These biomarkers have been shown to be potentially useful in multiple aspects of metastatic colorectal cancer, such as auxiliary diagnosis of metastasis, prognosis prediction, and monitoring of therapy response.
Collapse
Affiliation(s)
- Wei Gao
- Department of Internal Medicine-Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Yigui Chen
- Department of Internal Medicine-Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Jianwei Yang
- Department of Internal Medicine-Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Changhua Zhuo
- Department of Gastrointestinal Surgical Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Sha Huang
- Department of Internal Medicine-Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Hui Zhang
- Department of Hepatopancreatobiliary Surgical Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Yi Shi
- Department of Molecular Pathology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, China
| |
Collapse
|
87
|
Lv X, Zhang J, Zhang J, Guan W, Ren W, Liu Y, Xu G. A Negative Feedback Loop Between NAMPT and TGF-β Signaling Pathway in Colorectal Cancer Cells. Onco Targets Ther 2021; 14:187-198. [PMID: 33447060 PMCID: PMC7802777 DOI: 10.2147/ott.s282367] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/03/2020] [Indexed: 12/23/2022] Open
Abstract
Background Nicotinamide phosphoribosyltransferase (NAMPT) and the transforming growth factor-β (TGF-β) signaling pathway play important roles in colorectal tumorigenesis and progress. However, the underlying regulatory mechanisms between NAMPT and TGF-β signaling in colorectal cancer (CRC) remain poorly understood. Methods Public data were extracted from the Oncomine database and the PrognoScan database to investigate the mRNA expression and the prognostic value of NAMPT, respectively, in CRC. Western blot tests were performed to detect Smad2, Smad3, p-Smad2, p-Smad3, Smad4 expression in CRC cells transfected with human NAMPT-siRNA or NAMPT-overexpressing plasmid. TGF-β1 concentrations in culture supernatants were assayed using ELISA kits. The effect of TGF-β1 on NAMPT expression was evaluated by quantitative real-time PCR and Western blot. The dual-luciferase reporter assay was employed to confirm the binding of miR-1-3p to NAMPT 3ʹ-UTR. Subsequently, NAMPT levels in HCT116 cells transfected with the mimics and inhibitors of miR-1-3p were detected by quantitative real-time PCR and Western blot. Results NAMPT was overexpressed in human CRC and was correlated with short overall survival. NAMPT increased the protein expression levels of components in the TGF-β signaling pathway including Smad2, Smad3, and Smad4. Moreover, NAMPT promoted TGF-β1 secretion. Intriguingly, the TGF-β1 treatment down-regulated NAMPT expression at mRNA and protein levels in CRC cells which were partly through the up-regulation of miR-1-3p that directly bound to the NAMPT 3ʹ-UTR. These outcomes demonstrated that NAMPT was a downstream target of miR-1-3p and there was a negative association between NAMPT and miR-1-3p in CRC. Conclusion There is a negative feedback loop between NAMPT and the TGF-β signaling pathway in CRC cells, providing new insight into the mechanism underlying the regulatory pathways in CRC.
Collapse
Affiliation(s)
- Xiaoqun Lv
- Department of Pharmacy, Jinshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jinguo Zhang
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Jun Zhang
- Department of Pharmacy, Jinshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Wencai Guan
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Weifang Ren
- Department of Pharmacy, Jinshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Yujuan Liu
- Department of Pharmacy, Jinshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Guoxiong Xu
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
88
|
Koulouridi A, Messaritakis I, Gouvas N, Tsiaoussis J, Souglakos J. Immunotherapy in Solid Tumors and Gut Microbiota: The Correlation-A Special Reference to Colorectal Cancer. Cancers (Basel) 2020; 13:cancers13010043. [PMID: 33375686 PMCID: PMC7795476 DOI: 10.3390/cancers13010043] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/16/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Immunotherapy and immune checkpoint inhibitors have become the breakthrough treatment with extended responses and survival rates in various neoplasms. They use the immune system to defeat cancer, while gut microbiota seems to play a significant role in that attempt. To date, colorectal cancer patients have gained little benefit from immunotherapy. Only mismatch repair-deficient/microsatellite-unstable tumors seem to respond positively to immunotherapy. However, gut microbiota could be the key to expanding the use of immunotherapy to a greater range of colorectal cancer patients. In the current review study, the authors aimed to present and analyze the mechanisms of action and resistance of immunotherapy and the types of immune checkpoint inhibitors (ICIs) as well as their correlation to gut microbiota. A special reference will be made in the association of immunotherapy and gut microbiota in the colorectal cancer setting. Abstract Over the last few years, immunotherapy has been considered as a key player in the treatment of solid tumors. Immune checkpoint inhibitors (ICIs) have become the breakthrough treatment, with prolonged responses and improved survival results. ICIs use the immune system to defeat cancer by breaking the axes that allow tumors to escape immune surveillance. Innate and adaptive immunity are involved in mechanisms against tumor growth. The gut microbiome and its role in such mechanisms is a relatively new study field. The presence of a high microbial variation in the gut seems to be remarkably important for the efficacy of immunotherapy, interfering with innate immunity. Metabolic and immunity pathways are related with specific gut microbiota composition. Various studies have explored the composition of gut microbiota in correlation with the effectiveness of immunotherapy. Colorectal cancer (CRC) patients have gained little benefit from immunotherapy until now. Only mismatch repair-deficient/microsatellite-unstable tumors seem to respond positively to immunotherapy. However, gut microbiota could be the key to expanding the use of immunotherapy to a greater range of CRC patients.
Collapse
Affiliation(s)
- Asimina Koulouridi
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece;
| | - Ippokratis Messaritakis
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece;
- Correspondence: (I.M.); (J.S.); Tel.: +30-28-1039-4926 (I.M.); +30-28-1039-4712 (J.S.)
| | - Nikolaos Gouvas
- Medical School, University of Cyprus, 20537 Nicosia, Cyprus;
| | - John Tsiaoussis
- Department of Anatomy, School of Medicine, University of Crete, 70013 Heraklion, Greece;
| | - John Souglakos
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece;
- Department of Medical Oncology, University Hospital of Heraklion, 71110 Heraklion, Greece
- Correspondence: (I.M.); (J.S.); Tel.: +30-28-1039-4926 (I.M.); +30-28-1039-4712 (J.S.)
| |
Collapse
|
89
|
Brind'Amour A, Dubé P, Tremblay JF, Soucisse ML, Mack L, Bouchard-Fortier A, McCart JA, Govindarajan A, Bischof D, Haase E, Giacomantonio C, Hebbard P, Younan R, MacNeill A, Boulanger-Gobeil C, Sidéris L. Canadian guidelines on the management of colorectal peritoneal metastases. Curr Oncol 2020; 27:e621-e631. [PMID: 33380878 PMCID: PMC7755452 DOI: 10.3747/co.27.6919] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Modern management of colorectal cancer (crc) with peritoneal metastasis (pm) is based on a combination of cytoreductive surgery (crs), systemic chemotherapy, and hyperthermic intraperitoneal chemotherapy (hipec). Although the role of hipec has recently been questioned with respect to results from the prodige 7 trial, the role and benefit of a complete crs were confirmed, as observed with a 41-month gain in median survival in that study, and 15% of patients remaining disease-free at 5 years. Still, crc with pm is associated with a poor prognosis, and good patient selection is essential. Many questions about the optimal management approach for such patients remain, but all patients with pm from crc should be referred to, or discussed with, a pm surgical oncologist, because cure is possible. The objective of the present guideline is to offer a practical approach to the management of pm from crc and to reflect on the new practice standards set by recent publications on the topic.
Collapse
Affiliation(s)
- A Brind'Amour
- Department of Surgery, Hôpital Maisonneuve-Rosemont, Montreal
- Department of Surgery, chu de Québec-Université Laval, Quebec City
| | - P Dubé
- Department of Surgery, Hôpital Maisonneuve-Rosemont, Montreal
- Department of Surgery, University of Montreal, Montreal, QC
| | - J F Tremblay
- Department of Surgery, Hôpital Maisonneuve-Rosemont, Montreal
- Department of Surgery, University of Montreal, Montreal, QC
| | - M L Soucisse
- Department of Surgery, Hôpital Maisonneuve-Rosemont, Montreal
- Department of Surgery, University of Montreal, Montreal, QC
| | - L Mack
- Department of Surgery, University of Calgary, Calgary, AB
| | | | - J A McCart
- Department of Surgery, Mount Sinai Hospital, University of Toronto, Toronto, ON
- Department of Surgery, University of Toronto, Toronto, ON
| | - A Govindarajan
- Department of Surgery, Mount Sinai Hospital, University of Toronto, Toronto, ON
- Department of Surgery, University of Toronto, Toronto, ON
| | - D Bischof
- Department of Surgery, Mount Sinai Hospital, University of Toronto, Toronto, ON
- Department of Surgery, University of Toronto, Toronto, ON
| | - E Haase
- Department of Surgery, University of Alberta, Edmonton, AB
| | | | - P Hebbard
- Department of Surgery, University of Manitoba, Winnipeg, MB
| | - R Younan
- Department of Surgery, University of Montreal, Montreal, QC
- Department of Surgery, Centre Hospitalier de l'Université de Montréal, Montreal, QC
| | - A MacNeill
- Department of Surgery, University of British Columbia, Vancouver, BC
| | | | - L Sidéris
- Department of Surgery, Hôpital Maisonneuve-Rosemont, Montreal
- Department of Surgery, University of Montreal, Montreal, QC
| |
Collapse
|
90
|
Forster S, Radpour R. Molecular Immunotherapy: Promising Approach to Treat Metastatic Colorectal Cancer by Targeting Resistant Cancer Cells or Cancer Stem Cells. Front Oncol 2020; 10:569017. [PMID: 33240813 PMCID: PMC7680905 DOI: 10.3389/fonc.2020.569017] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/08/2020] [Indexed: 12/17/2022] Open
Abstract
The immune system is able to recognize and eliminate tumor cells. Some tumors, including colorectal cancer (CRC), induce immune tolerance via different mechanisms of “immunoediting” and “immune evasion” and can thus escape immune surveillance. The impact of immunotherapy on cancer has been investigated for many years, but so far, the application was limited to few cancer types. Immuno-oncological therapeutic strategies against metastatic colorectal cancer (mCRC), the adaptive immune system activating approaches, offer a high potential for adaptation to the great heterogeneity of CRC. Moreover, novel treatment approaches are currently being tested that might specifically target the disease initiating and maintaining population of colorectal cancer stem cells (CSCs). In this review, we aim to summarize the current state of immune-oncology and tumor immunotherapy of patients with mCRC and discuss different therapeutic modalities that focus on the activation of tumor-specific T-cells and their perspectives such as tumor vaccination, checkpoint inhibition, and adoptive T-cell transfer or on the eradication of colorectal CSCs.
Collapse
Affiliation(s)
- Stefan Forster
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ramin Radpour
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
91
|
Li HJ, Wang YL, Ming L, Guo XQ, Li YL, Wang JC, Zhang YQ, Cheng L. Development of a prognostic model based on an immunogenomic landscape analysis of colorectal cancer. Future Oncol 2020; 17:301-313. [PMID: 32996773 DOI: 10.2217/fon-2020-0022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Screening and therapeutic programs for colorectal cancer (CRC) are invasive or not effective and unable to meet patient needs. Major advances in immunogenomics may change this status but need more exploration. Differentially expressed genes and immune-related genes (IRGs) were identified by computational methods. A prognostic model was established and validated based on survival-related IRGs via stepwise multivariate Cox regression analysis. Nine IRGs were selected and identified as survival-related genes. A 7-gene prognostic model could offer a preliminary and valid determination of risk in CRC patients. The area under the curve of the receiver operating characteristic was 0.672. The 7-gene prognostic model might be used as a novel prognostic tool in CRC patients.
Collapse
Affiliation(s)
- H J Li
- The First Affiliated Hospital of Zhengzhou University, 450000, PR China.,Academy of Medical Science, Zhengzhou University, 450000, PR China
| | - Y L Wang
- The First Affiliated Hospital of Zhengzhou University, 450000, PR China.,Henan Bioengineering Research Center, 450100, PR China
| | - L Ming
- The First Affiliated Hospital of Zhengzhou University, 450000, PR China
| | - X Q Guo
- Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, 475000, PR China
| | - Y L Li
- Henan Bioengineering Research Center, 450100, PR China
| | - J C Wang
- Henan Bioengineering Research Center, 450100, PR China
| | - Y Q Zhang
- Henan Bioengineering Research Center, 450100, PR China.,Zhengzhou Technical College, 450100, PR China
| | - L Cheng
- Henan Bioengineering Research Center, 450100, PR China
| |
Collapse
|
92
|
Batra A, Rigo R, Hannouf MB, Cheung WY. Real-world Safety and Efficacy of Raltitrexed in Patients With Metastatic Colorectal Cancer. Clin Colorectal Cancer 2020; 20:e75-e81. [PMID: 33268287 DOI: 10.1016/j.clcc.2020.09.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/01/2020] [Accepted: 09/14/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Use of fluoropyrimidine-based therapy in patients with metastatic colorectal cancer is associated with significant toxicities. This study aimed to assess the safety and efficacy of raltitrexed use in patients with metastatic colorectal cancer who developed significant toxicities after fluoropyrimidine-based treatment. PATIENTS AND METHODS We identified patients with metastatic colorectal cancer who were treated with raltitrexed-based systemic therapy after developing serious adverse events with fluoropyrimidine-based treatment in a large Canadian province from 2004 to 2018. Demographic, tumor, and treatment characteristics were retrieved from the electronic medical records. Progression-free and overall survival were assessed from the start of raltitrexed-based therapy. RESULTS A total of 86 patients were identified for the study. The median age was 66.5 years, and 58.1% of patients were men. The primary cancer site was right, left, and transverse colon in 38.4%, 27.9%, and 9.3%, respectively. The remaining 24.4% had rectal cancer. Among all patients, 43.0% had received more than 2 prior systemic therapies, and 37.6% had developed previous cardiotoxicity to fluoropyrimidine-based treatment. The median progression-free and overall survival were 8.5 and 10.2 months, respectively. On multivariable Cox regression model, patients with left-sided colon cancer (hazard ratio [HR], 0.33; 95% confidence interval [CI], 0.12-0.97; P = .044) and the Eastern Cooperative Oncology Group performance status of 0/1 (HR, 0.10; 95% CI, 0.01-0.82; P = .032) had a longer progression-free survival, whereas left-sidedness of colon cancer was the only factor that predicted overall survival (HR, 0.30; 95% CI, 0.10-0.88; P = .029). Raltitrexed was well-tolerated with common adverse events that included anemia in 41.7% of patients and chemotherapy-induced nausea and vomiting in 27.4%. Most toxicities were grade 1/2, but 16.7% of patients experienced grade 3. There were no cardiac events and treatment-related deaths. CONCLUSIONS Raltitrexed in patients with colorectal cancer who were previously treated with fluoropyrimidine-based systemic therapy is effective and well-tolerated.
Collapse
Affiliation(s)
- Atul Batra
- Department of Medical Oncology, Tom Baker Cancer Center, Calgary, Alberta, Canada; University of Calgary, Calgary, Alberta, Canada
| | - Rodrigo Rigo
- Department of Medical Oncology, Tom Baker Cancer Center, Calgary, Alberta, Canada; University of Calgary, Calgary, Alberta, Canada
| | | | - Winson Y Cheung
- Department of Medical Oncology, Tom Baker Cancer Center, Calgary, Alberta, Canada; University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
93
|
Rizzo A, Mollica V, Cimadamore A, Santoni M, Scarpelli M, Giunchi F, Cheng L, Lopez-Beltran A, Fiorentino M, Montironi R, Massari F. Is There a Role for Immunotherapy in Prostate Cancer? Cells 2020; 9:E2051. [PMID: 32911806 PMCID: PMC7564598 DOI: 10.3390/cells9092051] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/04/2020] [Accepted: 09/06/2020] [Indexed: 02/06/2023] Open
Abstract
In the last decade, immunotherapy has revolutionized the treatment landscape of several hematological and solid malignancies, reporting unprecedented response rates. Unfortunately, this is not the case for metastatic castration-resistant prostate cancer (mCRPC), as several phase I and II trials assessing programmed death receptor 1 (PD-1) and cytotoxic T-lymphocyte antigen-4 (CTLA-4) inhibitors have shown limited benefits. Moreover, despite sipuleucel-T representing the only cancer vaccine approved by the Food and Drug Administration (FDA) for mCRPC following the results of the IMPACT trial, the use of this agent is relatively limited in everyday clinical practice. The identification of specific histological and molecular biomarkers that could predict response to immunotherapy represents one of the current challenges, with an aim to detect subgroups of mCRPC patients who may benefit from immune checkpoint monoclonal antibodies as monotherapy or in combination with other anticancer agents. Several unanswered questions remain, including the following: is there-or will there ever be-a role for immunotherapy in prostate cancer? In this review, we aim at underlining the failures and promises of immunotherapy in prostate cancer, summarizing the current state of art regarding cancer vaccines and immune checkpoint monoclonal antibodies, and discussing future research directions in this immunologically "cold" malignancy.
Collapse
Affiliation(s)
- Alessandro Rizzo
- Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, via Albertoni, 40138 Bologna, Italy; (A.R.); (V.M.)
| | - Veronica Mollica
- Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, via Albertoni, 40138 Bologna, Italy; (A.R.); (V.M.)
| | - Alessia Cimadamore
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy; (A.C.); (M.S.); (R.M.)
| | - Matteo Santoni
- Oncology Unit, Macerata Hospital, 62012 Macerata, Italy;
| | - Marina Scarpelli
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy; (A.C.); (M.S.); (R.M.)
| | - Francesca Giunchi
- Department of Pathology, Ospedale Maggiore and University of Bologna, 40138 Bologna, Italy; (F.G.); (M.F.)
| | - Liang Cheng
- Laboratory Medicine and Department of Pathology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | - Michelangelo Fiorentino
- Department of Pathology, Ospedale Maggiore and University of Bologna, 40138 Bologna, Italy; (F.G.); (M.F.)
| | - Rodolfo Montironi
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy; (A.C.); (M.S.); (R.M.)
| | - Francesco Massari
- Oncologia Medica, Azienda Ospedaliero-Universitaria di Bologna, via Albertoni, 40138 Bologna, Italy; (A.R.); (V.M.)
| |
Collapse
|
94
|
Wang H, Liu S, Kong F, Xiao F, Li Y, Wang H, Zhang S, Huang D, Wang L, Yang Y. Spred2 inhibits epithelial‑mesenchymal transition of colorectal cancer cells by impairing ERK signaling. Oncol Rep 2020; 44:174-184. [PMID: 32319644 PMCID: PMC7251656 DOI: 10.3892/or.2020.7586] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 03/16/2020] [Indexed: 12/31/2022] Open
Abstract
Downregulation of the sprouty-related EVH1 domain protein 2 (Spred2) is closely associated with highly metastatic phenotypes in various tumors. However, the roles of Spred2 in the development and progression of colorectal cancer (CRC) are still largely unexplored. As anticipated, Spred2 expression was significantly downregulated in clinical tumor tissues. To restore Spred2 levels, Ad.Spred2, an adenoviral vector expressing Spred2, was transduced into CRC cells. It was revealed that Ad.Spred2 inhibited the proliferation and decreased the survival and migration of SW480 cells. Epithelial-mesenchymal transition (EMT) is an essential event during tumor metastasis to distant sites. It was revealed that Ad.Spred2 markedly inhibited EMT by promoting F-actin reorganization, upregulating E-cadherin levels and reducing vimentin protein expression. Notably, extracellular-regulated kinase (ERK) signaling inhibition by PD98059 induced similar effects on EMT in CRC cells, indicating that Ad.Spred2 regulated EMT in CRC cells in an ERK-dependent manner. Transforming growth factor β (TGF-β), a well-known inducer of EMT, increased E-cadherin expression, decreased vimentin expression and promoted migration in CRC cells. However, neither Ad.Spred2 nor PD98059 had an obvious effect on the expression of SMAD2/3 or SMAD4 in SW480 cells, indicating that Ad.Spred2 inhibited EMT in a SMAD-independent manner. Notably, Ad.Spred2 transduction downregulated SAMD2/3 and SMAD4 levels in HCT116 cells in an ERK-independent manner. It was speculated that Ad.Spred2 inhibited the EMT of HCT116 cells by both blocking ERK signaling and reducing SMAD signaling. It was concluded that Spred2 inhibited EMT in CRC cells by interfering with ERK signaling, with or without reduced SMAD signaling. Therefore, the introduction of the clinical application of Spred2 has great potential for development as a gene therapy approach for CRC.
Collapse
Affiliation(s)
- Hao Wang
- Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Shuchen Liu
- Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Fanxuan Kong
- Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Fengjun Xiao
- Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Yuxiang Li
- Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Hua Wang
- Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Shun Zhang
- Department of Experimental Medical Science and Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315000, P.R. China
| | - Dandan Huang
- Department of Experimental Medical Science and Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang 315000, P.R. China
| | - Lisheng Wang
- Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Yuefeng Yang
- Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| |
Collapse
|
95
|
Hanna M, Guerra G, O'Byrne K. Durable complete response to immunotherapy in treatment-resistant metastatic colorectal cancer with thyroid transcription factor 1 expression. ANZ J Surg 2020; 90:E97-E99. [PMID: 32142186 DOI: 10.1111/ans.15810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 02/23/2020] [Indexed: 11/29/2022]
Affiliation(s)
- Mark Hanna
- Department of Colorectal Surgery, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Glen Guerra
- Department of Colorectal Surgery, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Kenneth O'Byrne
- Department of Medical Oncology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
96
|
Pan QZ, Gu JM, Zhao JJ, Tang Y, Wang QJ, Zhu Q, Song MJ, Li YQ, He J, Chen SP, Weng DS, Xia JC. Retrospective analysis of the efficacy of cytokine-induced killer cell immunotherapy combined with first-line chemotherapy in patients with metastatic colorectal cancer. Clin Transl Immunology 2020; 9:e1113. [PMID: 32076550 PMCID: PMC7029432 DOI: 10.1002/cti2.1113] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 02/03/2020] [Accepted: 02/03/2020] [Indexed: 12/14/2022] Open
Abstract
Objectives Fluoropyrimidine‐based chemotherapy regimens are the current first‐line treatment for metastatic colorectal cancer (mCRC); however, the outcome is often unsatisfactory. The present study aimed to determine the effect of combined cytokine‐induced killer (CIK) cell immunotherapy and first‐line chemotherapy in patients with mCRC. Methods This retrospective study included 252 patients with mCRC treated with first‐line chemotherapy. Among them, 126 patients received first‐line chemotherapy only (control group), while the other 126 patients, with similar demographic and clinical characteristics, received CIK cell immunotherapy combined with first‐line chemotherapy (CIK group). Overall survival (OS) and progression‐free survival (PFS) were compared between the two groups using the Kaplan–Meier method. Results The median OS for the CIK group was 54.7 versus 24.1 months for the controls, and the median PFS for the CIK group was 25.7 versus 14.6 months for the controls. Univariate and multivariate analyses indicated that CIK cell treatment was an independent prognostic factor for patients' OS and PFS. Subgroup analyses showed that CIK cell treatment significantly improved the OS and PFS of patients with metastatic colon cancer, but not those with metastatic rectal cancer. Additionally, the change in CD3+CD56+ subsets after the fourth treatment cycle might be an indicator of successful CIK cell treatment: Patients with increased CD3+CD56+ subsets had better survival than those with decreased CD3+CD56+ subsets. Conclusion Cytokine‐induced killer cell immunotherapy combined with first‐line chemotherapy could significantly improve the OS and PFS of patients with mCRC, particularly for patients with metastatic colon cancer.
Collapse
Affiliation(s)
- Qiu-Zhong Pan
- State Key Laboratory of Oncology in South China Collaborative Innovation Center for Cancer Medicine Sun Yat-Sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-Sen University Cancer Center Guangzhou China
| | - Jia-Mei Gu
- State Key Laboratory of Oncology in South China Collaborative Innovation Center for Cancer Medicine Sun Yat-Sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-Sen University Cancer Center Guangzhou China
| | - Jing-Jing Zhao
- State Key Laboratory of Oncology in South China Collaborative Innovation Center for Cancer Medicine Sun Yat-Sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-Sen University Cancer Center Guangzhou China
| | - Yan Tang
- State Key Laboratory of Oncology in South China Collaborative Innovation Center for Cancer Medicine Sun Yat-Sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-Sen University Cancer Center Guangzhou China
| | - Qi-Jing Wang
- Department of Biotherapy Sun Yat-Sen University Cancer Center Guangzhou China
| | - Qian Zhu
- State Key Laboratory of Oncology in South China Collaborative Innovation Center for Cancer Medicine Sun Yat-Sen University Cancer Center Guangzhou China
| | - Meng-Jia Song
- State Key Laboratory of Oncology in South China Collaborative Innovation Center for Cancer Medicine Sun Yat-Sen University Cancer Center Guangzhou China
| | - Yong-Qiang Li
- Department of Biotherapy Sun Yat-Sen University Cancer Center Guangzhou China
| | - Jia He
- Department of Biotherapy Sun Yat-Sen University Cancer Center Guangzhou China
| | - Shi-Ping Chen
- Department of Biotherapy Sun Yat-Sen University Cancer Center Guangzhou China
| | - De-Sheng Weng
- State Key Laboratory of Oncology in South China Collaborative Innovation Center for Cancer Medicine Sun Yat-Sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-Sen University Cancer Center Guangzhou China
| | - Jian-Chuan Xia
- State Key Laboratory of Oncology in South China Collaborative Innovation Center for Cancer Medicine Sun Yat-Sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-Sen University Cancer Center Guangzhou China
| |
Collapse
|
97
|
Kreidieh M, Mukherji D, Temraz S, Shamseddine A. Expanding the Scope of Immunotherapy in Colorectal Cancer: Current Clinical Approaches and Future Directions. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9037217. [PMID: 32090113 PMCID: PMC7008242 DOI: 10.1155/2020/9037217] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 08/16/2019] [Indexed: 12/14/2022]
Abstract
The success of immune checkpoint inhibitors (ICIs) in an increasing range of heavily mutated tumor types such as melanoma has culminated in their exploration in different subsets of patients with metastatic colorectal cancer (mCRC). As a result of their dramatic and durable response rates in patients with chemorefractory, mismatch repair-deficient-microsatellite instability-high (dMMR-MSI-H) mCRC, ICIs have become potential alternatives to classical systemic therapies. The anti-programmed death-1 (PD-1) agents, Pembrolizumab and Nivolumab, have been granted FDA approval for this subset of patients. Unfortunately, however, not all CRC cases with the dMMR-MSI-H phenotype respond well to ICIs, and ongoing studies are currently exploring biomarkers that can predict good response to them. Another challenge lies in developing novel treatment strategies for the subset of patients with the mismatch repair-proficient-microsatellite instability-low (pMMR-MSI-L) phenotype that comprises 95% of all mCRC cases in whom treatment with currently approved ICIs has been largely unsuccessful. Approaches aiming at overcoming the resistance of tumors in this subset of patients are being developed including combining different checkpoint inhibitors with either chemotherapy, anti-angiogenic agents, cancer vaccines, adoptive cell transfer (ACT), or bispecific T-cell (BTC) antibodies. This review describes the rationale behind using immunotherapeutics in CRC. It sheds light on the progress made in the use of immunotherapy in the treatment of patients with dMMR-MSI-H CRC. It also discusses emerging approaches and proposes potential strategies for targeting the immune microenvironment in patients with pMMR-MSI-L CRC tumors in an attempt to complement immune checkpoint inhibition.
Collapse
Affiliation(s)
- Malek Kreidieh
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | - Deborah Mukherji
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | - Sally Temraz
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali Shamseddine
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
98
|
Nompumelelo Simelane NW, Kruger CA, Abrahamse H. Photodynamic diagnosis and photodynamic therapy of colorectal cancer in vitro and in vivo. RSC Adv 2020; 10:41560-41576. [PMID: 35516575 PMCID: PMC9058000 DOI: 10.1039/d0ra08617g] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/08/2020] [Indexed: 02/06/2023] Open
Abstract
This review highlights the various photo diagnostic and treatment methods utilized for CRC, over the last seven years.
Collapse
Affiliation(s)
| | - Cherie Ann Kruger
- Laser Research Centre
- Faculty of Health Sciences
- University of Johannesburg
- Johannesburg 2028
- South Africa
| | - Heidi Abrahamse
- Laser Research Centre
- Faculty of Health Sciences
- University of Johannesburg
- Johannesburg 2028
- South Africa
| |
Collapse
|
99
|
Meng Y, Sun J, Qu N, Zhang G, Yu T, Piao H. Application of Radiomics for Personalized Treatment of Cancer Patients. Cancer Manag Res 2019; 11:10851-10858. [PMID: 31920394 PMCID: PMC6941598 DOI: 10.2147/cmar.s232473] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/16/2019] [Indexed: 12/14/2022] Open
Abstract
Radiomics is a novel concept that relies on obtaining image data from examinations such as computed tomography (CT), magnetic resonance imaging (MRI), or positron emission tomography (PET). With the appropriate algorithm, the extracted results have broad applicability and potential for a massive positive impact in radiology. For example, clinicians can verify treatment efficiency, predict the location of tumor metastasis, correlate results with a histopathological examination, or more accurately define the type of cancer. Combining radiomics with other testing techniques allows every patient to have a personalized treatment plan that is essential for advanced examination and treatment. This article explains the process of radiomics, including data collection mechanisms, combined use with genomics, and artificial intelligence and immunology techniques, which may solve many of the challenges faced by doctors in diagnosing and treating their patients.
Collapse
Affiliation(s)
- Yiming Meng
- Central Laboratory, Cancer Hospital of China Medical University, Liaoning Province Cancer Hospital, Shenyang 110042, People's Republic of China
| | - Jing Sun
- Central Laboratory, Cancer Hospital of China Medical University, Liaoning Province Cancer Hospital, Shenyang 110042, People's Republic of China
| | - Na Qu
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Province Cancer Hospital, Shenyang 110042, People's Republic of China
| | - Guirong Zhang
- Central Laboratory, Cancer Hospital of China Medical University, Liaoning Province Cancer Hospital, Shenyang 110042, People's Republic of China
| | - Tao Yu
- Department of Medical Image, Cancer Hospital of China Medical University, Liaoning Province Cancer Hospital, Shenyang 110042, People's Republic of China
| | - Haozhe Piao
- Central Laboratory, Cancer Hospital of China Medical University, Liaoning Province Cancer Hospital, Shenyang 110042, People's Republic of China.,Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Province Cancer Hospital, Shenyang 110042, People's Republic of China
| |
Collapse
|
100
|
Gessani S, Belardelli F. Immune Dysfunctions and Immunotherapy in Colorectal Cancer: The Role of Dendritic Cells. Cancers (Basel) 2019; 11:E1491. [PMID: 31623355 PMCID: PMC6827143 DOI: 10.3390/cancers11101491] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 02/08/2023] Open
Abstract
Colorectal cancer (CRC), a multi-step malignancy showing increasing incidence in today's societies, represents an important worldwide health issue. Exogenous factors, such as lifestyle, diet, nutrition, environment and microbiota, contribute to CRC pathogenesis, also influencing non neoplastic cells, including immune cells. Several immune dysfunctions were described in CRC patients at different disease stages. Many studies underline the role of microbiota, obesity-related inflammation, diet and host reactive cells, including dendritic cells (DC), in CRC pathogenesis. Here, we focused on DC, the main cells linking innate and adaptive anti-cancer immunity. Variations in the number and phenotype of circulating and tumor-infiltrating DC have been found in CRC patients and correlated with disease stages and progression. A critical review of DC-based clinical studies and of recent advances in cancer immunotherapy leads to consider new strategies for combining DC vaccination strategies with check-point inhibitors, thus opening perspectives for a more effective management of this neoplastic disease.
Collapse
Affiliation(s)
- Sandra Gessani
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | | |
Collapse
|