51
|
Zhou N, Tang Q, Yu H, Li T, Ren F, Zu L, Chen G, Chen J, Xu S. Comprehensive analyses of one-carbon metabolism related genes and their association with prognosis, tumor microenvironment, chemotherapy resistance and immunotherapy in lung adenocarcinoma. Front Mol Biosci 2022; 9:1034208. [PMID: 36438661 PMCID: PMC9699278 DOI: 10.3389/fmolb.2022.1034208] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/31/2022] [Indexed: 04/24/2024] Open
Abstract
Background: Lung adenocarcinoma (LUAD) is the most common type of lung cancer and is a global public health concern. One-carbon (1C) metabolism plays a crucial role in the occurrence and development of multiple cancer types. However, there are limited studies investigating 1C metabolism in LUAD. This study aims to evaluate the prognostic value of 1C metabolism-related genes in LUAD and to explore the potential correlation of these genes with gene methylation, the tumor microenvironment, and immunotherapy. Methods: We identified 26 1C metabolism-related genes and performed a Kaplan-Meier and Cox regression analysis to evaluate the prognostic value of these genes. Consensus clustering was further performed to determine the 1C metabolism-related gene patterns in LUAD. The clinical and molecular characteristics of subgroups were investigated based on consensus clustering. CIBERSORT and ssGSEA algorithms were used to calculate the relative infiltration levels of multiple immune cell subsets. The relationship between 1C metabolism-related genes and drug sensitivity to immunotherapy was evaluated using the CellMiner database and IMvigor210 cohort, respectively. Results: The expression levels of 23 1C metabolism-related genes were significantly different between LUAD tumor tissues and normal tissues. Seventeen of these genes were related to prognosis. Two clusters (cluster 1 and cluster 2) were identified among 497 LUAD samples based on the expression of 7 prognosis-related genes. Distinct expression patterns were observed between the two clusters. Compared to cluster 2, cluster 1 was characterized by inferior overall survival (OS) (median OS = 41 vs. 60 months, p = 0.00031), increased tumor mutation burden (15.8 vs. 7.5 mut/Mb, p < 0.001), high expression of PD-1 (p < 0.001) and PD-L1 (p < 0.001), as well as enhanced immune infiltration. 1C metabolism-related genes were positively correlated with the expression of methylation enzymes, and a lower methylation level was observed in cluster 1 (p = 0.0062). Patients in cluster 1 were resistant to chemotherapy drugs including pemetrexed, gemcitabine, paclitaxel, etoposide, oxaliplatin, and carboplatin. The specific expression pattern of 1C metabolism-related genes was correlated with a better OS in patients treated with immunotherapy (median OS: 11.2 vs. 7.8 months, p = 0.0034). Conclusion: This study highlights that 1C metabolism is correlated with the prognosis of LUAD patients and immunotherapy efficacy. Our findings provide novel insights into the role of 1C metabolism in the occurrence, development, and treatment of LUAD, and can assist in guiding immunotherapy for LUAD patients.
Collapse
Affiliation(s)
- Ning Zhou
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Quanying Tang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Haochuan Yu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Tong Li
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Fan Ren
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Lingling Zu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Gang Chen
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jun Chen
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Song Xu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
52
|
Zhu H, Wang X, Zhou X, Lu S, Gu G, Liu C. E3 ubiquitin ligase FBXW7 enhances radiosensitivity of non-small cell lung cancer cells by inhibiting SOX9 regulation of CDKN1A through ubiquitination. J Transl Med 2022; 102:1203-1213. [PMID: 36775446 DOI: 10.1038/s41374-022-00812-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/09/2022] [Accepted: 06/09/2022] [Indexed: 11/09/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) has high rates of morbidity and mortality. E3 ubiquitin ligase usually has antitumor effects. This study evaluated the mechanism of E3 ligase FBXW7 (F-box and WD repeat domain containing 7) in the radiosensitivity of NSCLC. NCI-H1299 and NCI-H1299R cells were irradiated by 0, 2, 4, and 6 Gy doses of X-ray, respectively. In addition to the measurement of cell proliferation, apoptosis, and γ-H2AX, FBXW7 expression was measured and the interaction between FBXW7 and SOX9 (SRY-box transcription factor 9) was evaluated. Ubiquitination level and protein stability of SOX9 were examined after FBXW7 overexpression. The binding relationship between SOX9 and CDKN1A (cyclin-dependent kinase inhibitor 1A) was verified. Xenograft tumor model was established to evaluate the effect of FBXW7 on radiosensitivity in vivo. FBXW7 was under-expressed in radioresistant cells. Overexpression of FBXW7 repressed NCI-H1299 and NCI-H1299R cell proliferation and colony formation and increased γ-H2AX-positive foci. Overexpression of FBXW7 increased the ubiquitination level and reduced the protein stability of SOX9. SOX9 bound to the CDKN1A promoter to inhibit CDKN1A expression. FBXW7 inhibited tumorigenesis and apoptosis and enhanced radiosensitivity of NSCLC cells in vivo via the SOX9/CDKN1A axis. Overall, FBXW7 inhibited SOX9 expression by promoting SOX9 ubiquitination and proteasome degradation, suppressing the binding of SOX9 to CDKN1A, and upregulating CDKN1A, thereby improving the radiosensitivity of NSCLC cells.
Collapse
Affiliation(s)
- Hongge Zhu
- The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Xiuli Wang
- The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Xin Zhou
- The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Suqiong Lu
- The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Guomin Gu
- The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Chunling Liu
- The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China.
| |
Collapse
|
53
|
Zu L, He J, Zhou N, Zeng J, Zhu Y, Tang Q, Jin X, Zhang L, Xu S. The Profile and Clinical Significance of ITGB2 Expression in Non-Small-Cell Lung Cancer. J Clin Med 2022; 11:6421. [PMID: 36362654 PMCID: PMC9655748 DOI: 10.3390/jcm11216421] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 10/26/2022] [Indexed: 08/13/2023] Open
Abstract
Integrins are involved in extracellular and intracellular signaling and are often aberrantly expressed in tumors. Integrin beta 2 (ITGB2) has previously been demonstrated to be correlated with the host defense. However, the expression profile and role of ITGB2 in non-small-cell lung cancer (NSCLC) remain unclear. Here, we found that the genetic alterations in ITGB2 was predominated by gene mutation and copy number deletion using cBioPortal analysis, and its expression was downregulated in the NSCLC tissues, as validated by the UALCAN, TCGA, and GEO databases and our tissue samples. Kaplan-Meier (KM) plotter analysis revealed that patients with a lower ITGB2 expression had a shorter overall survival (OS) time (p = 0.01). Moreover, 1089 differentially expressed genes (DEGs) in the NSCLC tissues were screened using the TCGA database. The GO and KEGG enrichment analysis showed that the DEGs were closely associated with immune processes and cell adhesion. The protein-protein interaction (PPI) network revealed that 10 of 15 EMT-related genes among the DEGs might lead to the metastasis of NSCLC. Concomitantly, the expression of ITGB2 was positively correlated with the infiltration of Treg cells and Myeloid-derived suppressor cells (MDSC). Biologically, the ectopic expression of ITGB2 significantly inhibited the proliferation and metastasis of NSCLC cells. Mechanistically, we demonstrated that ITGB2 suppressed the expression of N-cadherin, Vimentin, Slug, Snail, and Twist, while it promoted E-cadherin expression, according to gain-of-function studies. In conclusion, ITGB2 can inhibit the proliferation and migration of NSCLC cells, leading to a poor prognosis, via epithelial-mesenchymal transition (EMT) signaling.
Collapse
Affiliation(s)
- Lingling Zu
- School of Life Sciences, Tianjin University, Tianjin 300072, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jinling He
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ning Zhou
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jingtong Zeng
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yifang Zhu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Quanying Tang
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xin Jin
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Lei Zhang
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Song Xu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
54
|
Immunotherapy and Radiotherapy as an Antitumoral Long-Range Weapon-A Partnership with Unsolved Challenges: Dose, Fractionation, Volumes, Therapeutic Sequence. Curr Oncol 2022; 29:7388-7395. [PMID: 36290857 PMCID: PMC9601214 DOI: 10.3390/curroncol29100580] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/23/2022] Open
Abstract
Immunotherapy, the modern oncological treatment with immune checkpoint inhibitors (ICIs), has been part of the clinical practice for malignant melanoma for more than a decade. Anti-cytotoxic T-lymphocyte antigen 4 (CTLA4), anti-programmed cell death Protein 1 (PD-1), or anti programmed death-ligand 1 (PD-L1) agents are currently part of the therapeutic arsenal of metastatic or relapsed disease in numerous cancers; more recently, they have also been evaluated and validated as consolidation therapy in the advanced local stage. The combination with radiotherapy, a treatment historically considered loco-regional, changes the paradigm, offering-via synergistic effects-the potential to increase immune-mediated tumor destruction. However, the fragile balance between the tumoricidal effects through immune mechanisms and the immunosuppression induced by radiotherapy means that, in the absence of ICI, the immune-mediated potentiation effect of radiotherapy at a distance from the site of administration is rare. Through analysis of the preclinical and clinical data, especially the evidence from the PACIFIC clinical trial, we can consider that hypofractionated irradiation and reduction of the irradiated volume, in order to protect the immune-infiltrated tumor microenvironment, performed concurrently with the immunotherapy or a maximum of 2 weeks before the start of ICI treatment, could bring maximum benefits. In addition, avoiding radiation-induced lymphopenia (RILD) by protecting some anatomical lymphoid structures or large blood vessels, as well as the use of irradiation of partial tumor volumes, even in plurimetastatic disease, for the conversion of a "cold" immunological tumor into a "hot" immunological tumor are modern concepts of radiotherapy in the era of immunotherapy. Low-dose radiotherapy could also be proposed in plurimetastatic cases, the effect being different (modeling of the TME) from that of high doses per fraction irradiation (cell death with release of antigens that facilitates immune-mediated cell death).
Collapse
|
55
|
Qin Z, Yu G, Li R, Zhao J. Preparation of Triptolide Nano Drug Delivery System and Its Antitumor Activity In-Vitro. J Biomed Nanotechnol 2022. [DOI: 10.1166/jbn.2022.3442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Triptolide (as an effective antitumor drug) is limited in clinical application because of its poor solubility and absorption in-vivo. Herein, we prepared folic acid modified polymer micelles to encapsulate triptolide and enhance its biologicalavailability coupled with antitumor
effect. We prepared nano-micelles of triptolide through thin lipid film hydrational method. Physical properties and in vitro release characterization of Fol-Plla-cl-Peg-Plla-cl-Tmicelles were evaluated, while bioavailability of the formulation in rats was investigated. Tumor targeting
potential of micelles was determined by observing the uptake of A549 cells. In-Vitro antitumor activity of micelles and free triptolide (API) was investigated with MTT assay. The prepared polymer material exhibited no cytotoxicity. The particle size distribution of Fol-Plla-cl-Peg-Plla-cl-T
micelles was uniform and small, with good stability and high efficiency of entrapment. Triptolide In-Vitro release from micelles demonstrated slow and continuous released for 24 h. Compared with API, the half-life of micelles was prolonged, whilst its bioavailability in-vivo
was increased by about 6.35 times. More importantly, Fol-Plla-cl-Peg-Plla-cl-T micelles significantly improved the antitumor activity of triptolide and showed good tumor targeting potential. Fol-Plla-cl-Peg-Plla-cl-T micelles could improve the bioavailability and antitumor activity of triptolide,
amid demonstration of good tumor targeting and high safety.
Collapse
Affiliation(s)
- ZhongHua Qin
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Gusu District, Suzhou, 215006, China
| | - GuiPing Yu
- Department of Thoracic Surgery, Jiangyin People’s Hospital, Jiangyin, 214499, China
| | - Ran Li
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, China
| | - Jun Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Gusu District, Suzhou, 215006, China
| |
Collapse
|
56
|
Hu Y, Paris S, Bertolet G, Barsoumian HB, He K, Sezen D, Chen D, Wasley M, Silva JDA, Mitchell JA, Voss TA, Masrorpour F, Leyton CK, Yang L, Leuschner C, Puebla-Osorio N, Gandhi S, Nguyen QN, Cortez MA, Welsh JW. Combining a nanoparticle-mediated immunoradiotherapy with dual blockade of LAG3 and TIGIT improves the treatment efficacy in anti-PD1 resistant lung cancer. J Nanobiotechnology 2022; 20:417. [PMID: 36123677 PMCID: PMC9484155 DOI: 10.1186/s12951-022-01621-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/26/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND While improvements in immunoradiotherapy have significantly improved outcomes for cancer patients, this treatment approach has nevertheless proven ineffective at controlling the majority of malignancies. One of the mechanisms of resistance to immunoradiotherapy is that immune cells may be suppressed via the myriad of different immune checkpoint receptors. Therefore, simultaneous blockade of multiple immune checkpoint receptors may enhance the treatment efficacy of immunoradiotherapy. METHODS We combined NBTXR3-enhanced localized radiation with the simultaneous blockade of three different checkpoint receptors: PD1, LAG3, and TIGIT, and tested the treatment efficacy in an anti-PD1-resistant lung cancer model in mice. 129 Sv/Ev mice were inoculated with fifty thousand αPD1-resistant 344SQR cells in the right leg on day 0 to establish primary tumors and with the same number of cells in the left leg on day 4 to establish the secondary tumors. NBTXR3 was intratumorally injected into the primary tumors on day 7, which were irradiated with 12 Gy on days 8, 9, and 10. Anti-PD1 (200 µg), αLAG3 (200 µg), and αTIGIT (200 µg) were given to mice by intraperitoneal injections on days 5, 8, 11, 14, 21, 28, 35, and 42. RESULTS This nanoparticle-mediated combination therapy is effective at controlling the growth of irradiated and distant unirradiated tumors, enhancing animal survival, and is the only one that led to the destruction of both tumors in approximately 30% of the treated mice. Corresponding with this improved response is robust activation of the immune response, as manifested by increased numbers of immune cells along with a transcriptional signature of both innate and adaptive immunity within the tumor. Furthermore, mice treated with this combinatorial therapy display immunological memory response when rechallenged by the same cancer cells, preventing tumor engraftment. CONCLUSION Our results strongly attest to the efficacy and validity of combining nanoparticle-enhanced radiotherapy and simultaneous blockade of multiple immune checkpoint receptors and provide a pre-clinical rationale for investigating its translation into human patients.
Collapse
Affiliation(s)
- Yun Hu
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Sébastien Paris
- Department of Translational Science, Nanobiotix, Paris, France
| | - Genevieve Bertolet
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Hampartsoum B Barsoumian
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Kewen He
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Duygu Sezen
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Radiation Oncology, Koc University School of Medicine, Istanbul, Turkey
| | - Dawei Chen
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Mark Wasley
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Jordan DA Silva
- Department of Translational Science, Nanobiotix, Paris, France
| | - Joylise A Mitchell
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Tiffany A Voss
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Fatemeh Masrorpour
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Claudia Kettlun Leyton
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Liangpeng Yang
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Carola Leuschner
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Nahum Puebla-Osorio
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Saumil Gandhi
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Quynh-Nhu Nguyen
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Maria Angelica Cortez
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - James W Welsh
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| |
Collapse
|
57
|
Targeting prolyl isomerase Pin1 as a promising strategy to overcome resistance to cancer therapies. Pharmacol Res 2022; 184:106456. [PMID: 36116709 DOI: 10.1016/j.phrs.2022.106456] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/10/2022] [Accepted: 09/14/2022] [Indexed: 11/22/2022]
Abstract
The development of tumor therapeutic resistance is one of the important reasons for the failure of antitumor therapy. Starting with multiple targets and multiple signaling pathways is helpful in understanding the mechanism of tumor resistance. The overexpression of prolyl isomerase Pin1 is highly correlated with the malignancy of cancer, since Pin1 controls many oncogenes and tumor suppressors, as well as a variety of cancer-driving signaling pathways. Strikingly, numerous studies have shown that Pin1 is directly involved in therapeutic resistance. In this review, we mainly summarize the functions and mechanisms of Pin1 in therapeutic resistance of multifarious cancers, such as breast, liver, and pancreatic carcinomas. Furtherly, from the perspective of Pin1-driven cancer signaling pathways including Raf/MEK/ERK, PI3K/Akt, Wnt/β-catenin, NF-κB, as well as Pin1 inhibitors containing juglone, epigallocatechin-3-gallate (EGCG), all-trans retinoic acid (ATRA) and arsenic trioxide (ATO), it is better to demonstrate the important potential role and mechanism of Pin1 in resistance and sensitization to cancer therapies. It will provide new therapeutic approaches for clinical reversal and prevention of tumor resistance by employing synergistic administration of Pin1 inhibitors and chemotherapeutics, implementing combination therapy of Pin1-related cancer signaling pathway inhibitors and Pin1 inhibitors, and exploiting novel Pin1-specific inhibitors.
Collapse
|
58
|
Synergistic effects of radiotherapy and targeted immunotherapy in improving tumor treatment efficacy: a review. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2022; 24:2255-2271. [PMID: 35913663 DOI: 10.1007/s12094-022-02888-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 07/05/2022] [Indexed: 10/16/2022]
Abstract
Radiotherapy (RT), unlike chemotherapy, is one of the most routinely used and effective genotoxic and immune response inducing cancer therapies with an advantage of reduced side effects. However, cancer can relapse after RT owing to multiple factors, including acquired tumor resistance, immune suppressive microenvironment buildup, increased DNA repair, thus favoring tumor metastasis. Efforts to mitigate these undesirable effects have drawn interest in combining RT with immunotherapy, particularly the use of immune checkpoint inhibitors, to tilt the pre-existing tumor stromal microenvironment into long-lasting therapy-induced antitumor immunity at multiple metastatic sites (abscopal effects). This multimodal therapeutic strategy can alleviate the increased T cell priming and decrease tumor growth and metastasis, thus emerging as a significant approach to sustain as long-term antitumor immunity. To understand more about this synergism, a detailed cellular mechanism underlying the dynamic interaction between tumor and immune cells within the irradiated tumor microenvironment needs to be explored. Hence, in the present review, we have attempted to evaluate various RT-inducible immune factors, which can be targeted by immunotherapy and provide detailed explanation to optimally maximize their synergy with immunotherapy for long-lasting antitumor immunity. Moreover, we have critically assessed various combinatorial approaches along with their challenges and described strategies to modify them in addition to providing approaches for optimal synergistic effects of the combination.
Collapse
|
59
|
Zhang Z, Liu X, Chen D, Yu J. Radiotherapy combined with immunotherapy: the dawn of cancer treatment. Signal Transduct Target Ther 2022; 7:258. [PMID: 35906199 PMCID: PMC9338328 DOI: 10.1038/s41392-022-01102-y] [Citation(s) in RCA: 257] [Impact Index Per Article: 85.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/19/2022] [Accepted: 06/30/2022] [Indexed: 11/09/2022] Open
Abstract
Radiotherapy (RT) is delivered for purposes of local control, but can also exert systemic effect on remote and non-irradiated tumor deposits, which is called abscopal effect. The view of RT as a simple local treatment has dramatically changed in recent years, and it is now widely accepted that RT can provoke a systemic immune response which gives a strong rationale for the combination of RT and immunotherapy (iRT). Nevertheless, several points remain to be addressed such as the interaction of RT and immune system, the identification of the best schedules for combination with immunotherapy (IO), the expansion of abscopal effect and the mechanism to amplify iRT. To answer these crucial questions, we roundly summarize underlying rationale showing the whole immune landscape in RT and clinical trials to attempt to identify the best schedules of iRT. In consideration of the rarity of abscopal effect, we propose that the occurrence of abscopal effect induced by radiation can be promoted to 100% in view of molecular and genetic level. Furthermore, the “radscopal effect” which refers to using low-dose radiation to reprogram the tumor microenvironment may amplify the occurrence of abscopal effect and overcome the resistance of iRT. Taken together, RT could be regarded as a trigger of systemic antitumor immune response, and with the help of IO can be used as a radical and systemic treatment and be added into current standard regimen of patients with metastatic cancer.
Collapse
Affiliation(s)
- Zengfu Zhang
- Department of Radiation Oncology, Shandong University Cancer Center, Yantai Road, No. 2999, Jinan, Shandong, China
| | - Xu Liu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jiyan Road, No. 440, Jinan, Shandong, China
| | - Dawei Chen
- Department of Radiation Oncology, Shandong University Cancer Center, Yantai Road, No. 2999, Jinan, Shandong, China.
| | - Jinming Yu
- Department of Radiation Oncology, Shandong University Cancer Center, Yantai Road, No. 2999, Jinan, Shandong, China.
| |
Collapse
|
60
|
Lee W, Kim S, An J, Kim TK, Cha H, Chang H, Kim S, Kim S, Han M. Tristetraprolin regulates phagocytosis through interaction with CD47 in head and neck cancer. Exp Ther Med 2022; 24:541. [PMID: 35978923 PMCID: PMC9366311 DOI: 10.3892/etm.2022.11478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 05/03/2022] [Indexed: 11/17/2022] Open
Abstract
CD47 is expressed in all human cancer cells, including head and neck cancer, and initiates a signaling cascade to inhibit macrophage phagocytosis. However, the mechanism underlying CD47 overexpression has not been elucidated in radioresistant head and neck cancer. The present study demonstrated that decreased Tristetraprolin (TTP) expression induced a sustained overexpression of CD47 using reverse transcription-quantitative PCR and western blotting, and that CD47 overexpression prevented phagocytosis using a phagocytosis assay in a radioresistant HN31R cell line. Subsequently, using TTP transfection, RNA interference, duel-luciferase assay and EMSA, it was revealed that TTP transfection enhanced phagocytosis through degradation of CD47 mRNA by directly binding to CD47 AREs within the CD47 3'UTR. Based on our previous study, methylation-specific PCR and western blotting revealed that DNMT1 was overexpressed in radioresistant HN31R cell line and TTP expression was decreased epigenetically by DMNT1 associated DNA methylation. Overall, these findings provided novel insight into the role of TTP as a biomarker of CD47-positive head and neck cancer patients.
Collapse
Affiliation(s)
- Won Lee
- Department of Otolaryngology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Republic of Korea
| | - Song Kim
- Department of Otolaryngology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Republic of Korea
| | - Jae An
- Department of Otolaryngology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Republic of Korea
| | - Tae-Koon Kim
- Department of Otolaryngology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Republic of Korea
| | - Hee Cha
- Department of Pathology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Republic of Korea
| | - Hyo Chang
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Sang Kim
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Seong Kim
- Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Myung Han
- Department of Otolaryngology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Republic of Korea
| |
Collapse
|
61
|
Wen X, Zeng X, Cheng X, Zeng X, Liu J, Zhang Y, Li Y, Chen H, Huang J, Guo Z, Chen X, Zhang X. PD-L1-Targeted Radionuclide Therapy Combined with αPD-L1 Antibody Immunotherapy Synergistically Improves the Antitumor Effect. Mol Pharm 2022; 19:3612-3622. [PMID: 35652897 DOI: 10.1021/acs.molpharmaceut.2c00281] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Immune checkpoint blockers (ICBs) targeting programmed death receptor 1 (PD-1) ligand 1 (PD-L1) for immunotherapy have radically reformed oncology. It is of great significance to enhance the response rate of ICB in cancer patients. Here, a radioiodinated anti-PD-L1 antibody (131I-αPD-L1) was developed for PD-L1-targeted single-photon emission computed tomography (SPECT) imaging and αPD-L1 immunotherapy. Flow cytometry and immunofluorescence staining were performed to identify PD-L1 upregulation in a time- and dose-dependent manner after being induced by 131I-αPD-L1. ImmunoSPECT imaging and biodistributions of 131I-αPD-L1 in CT26, MC38, 4T1, and B16F10 tumor models were conducted to visualize the high tumor uptake and low background signal. Compared to monotherapy alone, concurrent administration of αPD-L1 mAb and 131I-αPD-L1 revealed improved tumor control in murine tumor models. The combination of 11.1 MBq of 131I-αPD-L1 and 200 μg of αPD-L1 mAb resulted in significant tumor growth delay and prolonged survival. This radioligand synergized immunotherapy strategy holds great potential for cancer management.
Collapse
Affiliation(s)
- Xuejun Wen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Xueyuan Zeng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Xingxing Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Xinying Zeng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Jia Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Yiren Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Yesen Li
- Department of Nuclear Medicine & Minnan PET Center, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Haojun Chen
- Department of Nuclear Medicine & Minnan PET Center, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Jinxiong Huang
- Department of Nuclear Medicine & Minnan PET Center, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Zhide Guo
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology and Surgery, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore 119074, Singapore.,Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.,Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.,Departments of Chemical and Biomolecular Engineering, and Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore 119074, Singapore
| | - Xianzhong Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen 361102, China
| |
Collapse
|
62
|
Shang Q, Dong Y, Su Y, Leslie F, Sun M, Wang F. Local scaffold-assisted delivery of immunotherapeutic agents for improved cancer immunotherapy. Adv Drug Deliv Rev 2022; 185:114308. [PMID: 35472398 DOI: 10.1016/j.addr.2022.114308] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 12/18/2022]
Abstract
Cancer immunotherapy, which reprograms a patient's own immune system to eradicate cancer cells, has been demonstrated as a promising therapeutic strategy clinically. Immune checkpoint blockade (ICB) therapies, cytokine therapies, cancer vaccines, and chimeric antigen receptor (CAR) T cell therapies utilize immunotherapy techniques to relieve tumor immune suppression and/or activate cellular immune responses to suppress tumor growth, metastasis and recurrence. However, systemic administration is often hampered by limited drug efficacy and adverse side effects due to nonspecific tissue distribution of immunotherapeutic agents. Advancements in local scaffold-based delivery systems facilitate a controlled release of therapeutic agents into specific tissue sites through creating a local drug reservoir, providing a potent strategy to overcome previous immunotherapy limitations by improving site-specific efficacy and minimizing systemic toxicity. In this review, we summarized recent advances in local scaffold-assisted delivery of immunotherapeutic agents to reeducate the immune system, aiming to amplify anticancer efficacy and minimize immune-related adverse events. Additionally, the challenges and future perspectives of local scaffold-assisted cancer immunotherapy for clinical translation and applications are discussed.
Collapse
Affiliation(s)
- Qi Shang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Yabing Dong
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China
| | - Yun Su
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China; Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21231, United States
| | - Faith Leslie
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD 21218, United States; Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, United States
| | - Mingjiao Sun
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, The Johns Hopkins University, Baltimore, MD 21218, United States; Institute for NanoBiotechnology, The Johns Hopkins University, Baltimore, MD 21218, United States; Department of Ophthalmology, School of Medicine, The Johns Hopkins University, Baltimore, MD 21231, United States
| | - Feihu Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| |
Collapse
|
63
|
Li J, Sun J, Liu Z, Zeng Z, Ouyang S, Zhang Z, Ma M, Kang W. The Roles of Non-Coding RNAs in Radiotherapy of Gastrointestinal Carcinoma. Front Cell Dev Biol 2022; 10:862563. [PMID: 35517505 PMCID: PMC9065280 DOI: 10.3389/fcell.2022.862563] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/22/2022] [Indexed: 12/19/2022] Open
Abstract
Radiotherapy (RT), or radiation therapy, has been widely used in clinical practice for the treatment of local advanced gastrointestinal carcinoma. RT causes DNA double-strand breaks leading to cell cytotoxicity and indirectly damages tumor cells by activating downstream genes. Non-coding RNA (including microRNAs, long non-coding RNAs (ncRNAs), and circular RNAs) is a type of RNA that does not encode a protein. As the field of ncRNAs increasingly expands, new complex roles have gradually emerged for ncRNAs in RT. It has been shown that ncRNAs can act as radiosensitivity regulators in gastrointestinal carcinoma by affecting DNA damage repair, cell cycle arrest, irradiation-induced apoptosis, cell autophagy, stemness, EMT, and cell pyroptosis. Here, we review the complex roles of ncRNAs in RT and gastrointestinal carcinoma. We also discuss the potential clinical significance and predictive value of ncRNAs in response to RT for guiding the individualized treatment of patients. This review can serve as a guide for the application of ncRNAs as radiosensitivity enhancers, radioresistance inducers, and predictors of response in RT of gastrointestinal carcinoma.
Collapse
|
64
|
Wu X, Wu J, Wang L, Yang W, Wang B, Yang H. CircRNAs in Malignant Tumor Radiation: The New Frontier as Radiotherapy Biomarkers. Front Oncol 2022; 12:854678. [PMID: 35372031 PMCID: PMC8966018 DOI: 10.3389/fonc.2022.854678] [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: 01/14/2022] [Accepted: 02/18/2022] [Indexed: 12/14/2022] Open
Abstract
World Health Organization (WHO) data show that of the top 20 factors that threaten human life and health, cancer is at the forefront, and the therapeutic approaches for cancer consist of surgery, radiotherapy, chemotherapy and immunotherapy. For most highly metastatic and recurrent cancer, radiation therapy is an essential modality to mitigate tumor burden and improve patient survival. Despite the great accomplishments that have been made in clinical therapy, an inevitable challenge in effective treatment is radioresistance, the mechanisms of which have not yet been completely elucidated. In addition, radiosensitization methods based on molecular mechanisms and targets, and clinical applications are still inadequate. Evidence indicates that circular RNAs (circRNAs) are important components in altering tumor progression, and in influencing resistance and susceptibility to radiotherapy. This review summarizes the reasons for tumor radiotherapy resistance induced by circRNAs, and clarifies the molecular mechanisms and targets of action. Moreover, we determine the potential value of circRNAs as clinical indicators in radiotherapy, providing a theoretical basis for circRNAs-based strategies for cancer radiotherapy.
Collapse
Affiliation(s)
- Xixi Wu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Junying Wu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Department of Clinical Laboratory, The Children's Hospital of Soochow University, Suzhou, China
| | - Lingxia Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei Yang
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Bo Wang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Huan Yang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China.,State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| |
Collapse
|
65
|
Yin G, Guo W, Huang Z, Chen X. Efficacy of radiotherapy combined with immune checkpoint inhibitors in patients with melanoma: a systemic review and meta-analysis. Melanoma Res 2022; 32:71-78. [PMID: 35254329 DOI: 10.1097/cmr.0000000000000800] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The purpose of this study is to review the efficacy of radiotherapy combined with immune checkpoint inhibitors (ICIs) in the treatment of melanoma and systematically evaluate the efficacy and safety of this combined treatment compared with ICIs alone. We searched a number of online databases up to 1 July 2021. Comprehensive Meta-Analysis 2.0 and RevMan 5.0 were used for summary analysis. The overall survival (OS), progression-free survival (PFS), overall response rate (ORR) and treatment adverse effects (AEs) were calculated. In total, 624 patients were included from 12 studies, including nine published studies and the results of three clinical trials. Radiotherapy combined with ICIs had a higher ORR compared with ICIs alone (35.00 vs. 20.39%). In terms of survival effect, radiotherapy combined with ICIs had no obvious advantage in OS. There was no statistically significant difference between 6-month and 12-month OS (P = 0.13; P = 0.69). There was no significant difference in PFS at 6 months (P = 0.08), but there was a significant difference in PFS at 12 months (P = 0.005). For patients with melanoma, radiotherapy combined with ICIs can improve the effective rate of treatment. Although there is no obvious OS advantage, it can improve PFS without serious adverse effects. Most of the studies included in this article are retrospective analyses, and there are few randomized controlled studies at present. Therefore, more prospective studies are still needed to explore the efficacy of radiotherapy combined with immunotherapy in melanoma.
Collapse
Affiliation(s)
- Gaofei Yin
- Department of Otorhinolaryngology and Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | | | | | | |
Collapse
|
66
|
Dietary Phytochemicals Targeting Nrf2 to Enhance the Radiosensitivity of Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7848811. [PMID: 35368867 PMCID: PMC8967572 DOI: 10.1155/2022/7848811] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/18/2022] [Accepted: 03/11/2022] [Indexed: 12/15/2022]
Abstract
Nowadays, cancer has become the second leading cause of death worldwide. Radiotherapy (RT) is the mainstay in management of carcinoma; however, overcoming radioresistance remains a great challenge to successfully treat cancer. Nrf2 is a key transcription factor that is responsible for maintaining cellular redox homeostasis. Activation of Nrf2 signaling pathway could upregulate multifarious antioxidant and detoxifying enzymes, further scavenging excessive reactive oxygen species (ROS). Despite its cytoprotective roles in normal cells, it could also alleviate oxidative stress and DNA damage caused by RT in cancer cells, thus promoting cancer cell survival. Accumulating evidence indicates that overactivation of Nrf2 is associated with radioresistance; therefore, targeting Nrf2 is a promising strategy to enhance radiosensitivity. Dietary phytochemicals coming from natural products are characterized by low cost, low toxicity, and general availability. Numerous phytochemicals are reported to regulate Nrf2 and intensify the killing capability of RT through diverse mechanisms, including promoting oxidative stress, proapoptosis, and proautophagy as well as inhibiting Nrf2-mediated cytoprotective genes expression. This review summarizes recent advances in radiosensitizing effects of dietary phytochemicals by targeting Nrf2 and discusses the underlying mechanisms, including N6-methyladenosine (m6A) modification of Nrf2 mediated by phytochemicals in cancer.
Collapse
|
67
|
Wang J, Zhuo LG, Zhao P, Liao W, Wei H, Yang Y, Peng S, Yang X. Screening for a 177Lu-labeled CA19-9 monoclonal antibody via PET imaging for colorectal cancer therapy. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.03.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
68
|
Khan SY, Melkus MW, Rasha F, Castro M, Chu V, Brandi L, Khan H, Gill HS, Pruitt K, Layeequr Rahman R. Tumor-Infiltrating Lymphocytes (TILs) as a Biomarker of Abscopal Effect of Cryoablation in Breast Cancer: A Pilot Study. Ann Surg Oncol 2022; 29:2914-2925. [PMID: 35094188 PMCID: PMC8990945 DOI: 10.1245/s10434-021-11157-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 11/14/2021] [Indexed: 12/29/2022]
Abstract
Abstract
Background
Morphological evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer is gaining momentum as an immunological biomarker. This experiment evaluates the role of TILs in distant tumors as a measure of abscopal effect from cryoablation of breast cancer.
Methods
BALB/c mice underwent bilateral orthotopic transplant with 4T1-12B (triple-negative) cells. At 2 weeks, left tumors were treated by either resection (standard of care group) or cryoablation (intervention group) followed by resection of the distant right tumors 1 week posttreatment. TIL scores were calculated from hematoxylin and eosin-stained sections and phenotyped for cytotoxic T-lymphocyte (CTL) markers by immunofluorescence. Primarily resected tumors served as baseline (Tbaseline), whereas resected distant right-sided served as the readout for abscopal effect (AbsRes or AbsCryo). Mice were monitored for tumor recurrence and metastasis.
Results
The AbsCryo had a significant mean (SD) increase in stromal (2.8 [1.1]%; p = 0.015) and invasive margin TILs (50 [12]%; p = 0.02) compared with TBaseline (1.0 [0]% and 31 [4.9]%, respectively). CTL phenotyping revealed a significant increase in mean (SD) CD8+ T cells (15.7 [12.1]; p = 0.02) and granzyme B (4.8 [3.6]; p = 0.048) for the AbsCryo compared with TBaseline (5.2 [4.7] and 2.4 [0.9], respectively). Posttreatment, the cryoablation group had no recurrence or metastasis, whereas the resected group showed local recurrence and lung metastasis in 40% of the mice. Postprocedure increase in TIL score of distant tumors was associated with decrease in tumor relapse (p = 0.02).
Conclusions
Cryoablation induced a robust tumor-specific TIL response compared with resection, suggesting an abscopal effect leading to the prevention of cancer recurrence and metastasis.
Collapse
Affiliation(s)
- Sonia Y Khan
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX, 79430-8312, USA
| | - Michael W Melkus
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX, 79430-8312, USA
| | - Fahmida Rasha
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Maribel Castro
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX, 79430-8312, USA
| | - Victoria Chu
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX, 79430-8312, USA
| | - Luis Brandi
- Department of Pathology, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Hafiz Khan
- Department of Public Health, Julia Jones Matthews, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Harvinder Singh Gill
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX, 79409, USA
| | - Kevin Pruitt
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Rakhshanda Layeequr Rahman
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX, 79430-8312, USA.
- Breast Center of Excellence, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
69
|
Ren J, Li L, Yu B, Xu E, Sun N, Li X, Xing Z, Han X, Cui Y, Wang X, Zhang X, Wang G. Extracellular vesicles mediated proinflammatory macrophage phenotype induced by radiotherapy in cervical cancer. BMC Cancer 2022; 22:88. [PMID: 35062905 PMCID: PMC8781113 DOI: 10.1186/s12885-022-09194-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 01/11/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Radiotherapy is a highly effective treatment for cervical cancer. Recent studies focused on the radiotherapy induced anti-tumor immunity. Whether tumor-derived extracellular vesicles (EVs) play roles in radiotherapy induced tumor associated macrophage (TAM) polarization remains unclear. MATERIALS AND METHODS This study analysed the phenotype of macrophages in cancer tissue and peripheral blood of cervical cancer patients using flow cytometry analysis. The role of EVs from plasma of post-irradiated patients on M2-like transformed macrophages was assessed. The M1- and M2-like macrophages were assessed by expression of cell surface markers (CCR7, CD163) and intracellular cytokines (IL-10, TNFα and iNOS). The capacity of phagocytosis was assessed by PD-1 expression and phagocytosis of pHrodo Red E. coli bioparticles. RESULTS Our results demonstrated that radiotherapy of cervical cancer induced an increase in the number of TAMs and a change in their subtype from the M2-like to the M1-like phenotype (increased expression of CCR7 and decreased expression of CD163). The EVs from plasma of post-irradiated patients facilitated the M2-like to the M1-like phenotype transition (increased expression of CCR7, TNFα and iNOS, and decreased expression of CD163 and IL-10) and increased capacity of phagocytosis (decreased PD-1 expression and increased phagocytosis of pHrodo Red E. coli bioparticles). CONCLUSIONS Our data demonstrated that irradiation in cervical cancer patients facilitated a proinflammatory macrophage phenotype which could eventually able to mediate anti-tumor immune responses. Our findings highlight the importance of EV in the crosstalk of tumor cells and TAM upon irradiation, which potentially leading to an increased inflammatory response to cancer lesions.
Collapse
Affiliation(s)
- Junli Ren
- Department of Radiotherapy Abdominopelvic, Shanxi Cancer Hospital, Taiyuan, 030013, Shanxi, China.
| | - Lili Li
- Department of Radiotherapy Abdominopelvic, Shanxi Cancer Hospital, Taiyuan, 030013, Shanxi, China
| | - Baofeng Yu
- Department of Biochemistry and Molecular biology, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Enwei Xu
- Department of Pathology, Shanxi Cancer Hospital, Taiyuan, 030013, Shanxi, China
| | - Naiping Sun
- Department of Radiotherapy Abdominopelvic, Shanxi Cancer Hospital, Taiyuan, 030013, Shanxi, China
| | - Xiaoning Li
- Department of Biochemistry and Molecular biology, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Zihan Xing
- Department of Biochemistry and Molecular biology, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Xiaodong Han
- Department of Radiotherapy Abdominopelvic, Shanxi Cancer Hospital, Taiyuan, 030013, Shanxi, China
| | - Yaqin Cui
- Department of Radiotherapy Abdominopelvic, Shanxi Cancer Hospital, Taiyuan, 030013, Shanxi, China
| | - Xiaoyan Wang
- Department of Gynecology, Shanxi Cancer Hospital, Taiyuan, 030013, Shanxi, China
| | - Xiaoxue Zhang
- Department of Pediatric Surgery, Xiang'an Hospital of Xiamen University, Xiamen, 350213, Fujian, China.
| | - Guoliang Wang
- National Center for Children's Health (NCCH), Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China.
| |
Collapse
|
70
|
Li K, Zhang Z, Mei Y, Li M, Yang Q, WU Q, Yang H, HE LIANGCAN, Liu S. Targeting innate immune system by nanoparticles for cancer immunotherapy. J Mater Chem B 2022; 10:1709-1733. [DOI: 10.1039/d1tb02818a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Various cancer therapies have advanced remarkably over the past decade. Unlike the direct therapeutic targeting of tumor cells, cancer immunotherapy is a new strategy that boosts the host's immune system...
Collapse
|
71
|
Analysis of the Dose Drop at the Edge of the Target Area in Heavy Ion Radiotherapy. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:4440877. [PMID: 34804193 PMCID: PMC8601813 DOI: 10.1155/2021/4440877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 10/13/2021] [Accepted: 10/15/2021] [Indexed: 11/30/2022]
Abstract
Background The dose distribution of heavy ions at the edge of the target region will have a steep decay during radiotherapy, which can better protect the surrounding organs at risk. Objective To analyze the dose decay gradient at the back edge of the target region during heavy ion radiotherapy. Methods Treatment planning system (TPS) was employed to analyze the dose decay at the edge of the beam under different incident modes and multiple dose segmentation conditions during fixed beam irradiation. The dose decay data of each plan was collected based on the position where the rear edge of the beam began to fall rapidly. Uniform scanning mode was selected in heavy ion TPS. Dose decay curves under different beam setup modes were drawn and compared. Results The dose decay data analysis showed that in the case of single beam irradiation, the posterior edge of the beam was 5 mm away, and the posterior dose could drop to about 20%. While irradiation in opposite direction, the posterior edge of the beam was 5 mm away, and the dose could drop to about 50%. In orthogonal irradiation of two beams, the posterior edge of the beam could drop to about 30-38% in a distance of 5 mm. Through the data analysis in the TPS, the sharpness of the dose at the back edge of the heavy ion beam is better than that at the lateral edge, but the generated X-ray contamination cannot be ignored. Conclusions The effect of uneven CT value on the dose decay of heavy ion beam should also be considered in clinical treatment.
Collapse
|
72
|
Gkika E, Adebahr S, Brenner A, Schimek-Jasch T, Radicioni G, Exner JP, Rühle A, Spohn SKB, Popp I, Zamboglou C, Sprave T, Firat E, Niedermann G, Nicolay NH, Nestle U, Grosu AL, Duda DG. Changes in Blood Biomarkers of Angiogenesis and Immune Modulation after Radiation Therapy and Their Association with Outcomes in Thoracic Malignancies. Cancers (Basel) 2021; 13:cancers13225725. [PMID: 34830880 PMCID: PMC8616228 DOI: 10.3390/cancers13225725] [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: 10/25/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022] Open
Abstract
The effects of radiotherapy on systemic immunity remain to be fully characterized in a disease-specific manner. The aim of the study was to examine potential biomarkers of systemic immunomodulation when using radiotherapy for thoracic malignancies. Serial blood samples were collected from 56 patients with thoracic malignancies prior (RTbaseline), during (RTduring) and at the end of radiotherapy (RTend), as well as at the first (FU1) and second follow-up (FU2). The changes in serum levels of IL-10, IFN-γ, IL-12p70, IL-13, IL-1β, IL-4, IL-6, IL-8, TNF-α, bFGF, sFlt-1, PlGF, VEGF, VEGF-C, VEGF-D and HGF were measured by multiplexed array and tested for associations with clinical outcomes. We observed an increase in the levels of IL-10, IFN-γ, PlGF and VEGF-D and a decrease in those of IL-8, VEGF, VEGF-C and sFlt-1 during and at the end of radiotherapy. Furthermore, baseline concentration of TNF-α significantly correlated with OS. IL-6 level at RTend and FU1,2 correlated with OS (RTend: p = 0.039, HR: 1.041, 95% CI: 1.002-1.082, FU1: p = 0.001, HR: 1.139, 95% CI: 1.056-1.228, FU2: p = 0.017, HR: 1.101 95% CI: 1.018-1.192), while IL-8 level correlated with OS at RTduring and RTend (RTduring: p = 0.017, HR: 1.014, 95% CI: 1.002-1.026, RTend: p = 0.004, HR: 1.007, 95% CI: 1.061-1.686). In conclusion, serum levels of TNF-α, IL-6 and IL-8 are potential biomarkers of response to radiotherapy. Given the recent implementation of immunotherapy in lung and esophageal cancer, these putative blood biomarkers should be further validated and evaluated in the combination or sequential therapy setting.
Collapse
Affiliation(s)
- Eleni Gkika
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Correspondence:
| | - Sonja Adebahr
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Anton Brenner
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
| | - Tanja Schimek-Jasch
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Gianluca Radicioni
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Jan-Philipp Exner
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Alexander Rühle
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Simon K. B. Spohn
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Ilinca Popp
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Constantinos Zamboglou
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Tanja Sprave
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Elke Firat
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Gabriele Niedermann
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Nils Henrik Nicolay
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Ursula Nestle
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- Department of Radiation Oncology, Kliniken Maria Hilf, 41063 Moenchengladbach, Germany
| | - Anca-Ligia Grosu
- University Medical Center Freiburg, Department of Radiation Oncology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (S.A.); (A.B.); (T.S.-J.); (G.R.); (J.-P.E.); (A.R.); (S.K.B.S.); (I.P.); (C.Z.); (T.S.); (E.F.); (G.N.); (N.H.N.); (U.N.); (A.-L.G.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Dan G. Duda
- E. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA;
| |
Collapse
|
73
|
Radiation-Induced Fibrotic Tumor Microenvironment Regulates Anti-Tumor Immune Response. Cancers (Basel) 2021; 13:cancers13205232. [PMID: 34680381 PMCID: PMC8533839 DOI: 10.3390/cancers13205232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 01/04/2023] Open
Abstract
Simple Summary Radiation therapy can modulate anti-tumor immune responses. In this study, we investigated the relationship between the anti-tumor immune response and tumor fibrosis after X-ray or neutron radiation therapy. Neutron radiation therapy resulted in lesser fibrosis and greater anti-tumor immunity compared to X-ray irradiation. Radiation therapy-induced fibrotic changes within the tumor environment and tumor regrowth were suppressed by specifically deleting Trp53 in endothelial cells. In particular, the upregulation of PD-L1 expression after X-ray radiation therapy was significantly suppressed via EC-Trp53 deletion. Understanding the effects of different radiation therapy types on the tumor microenvironment provides strategies for enhancing the efficacy of combined radio- and immunotherapy. Abstract High linear energy transfer (LET) radiation, such as neutron radiation, is considered more effective for the treatment of cancer than low LET radiation, such as X-rays. We previously reported that X-ray irradiation induced endothelial-to-mesenchymal transition (EndMT) and profibrotic changes, which contributed to the radioresistance of tumors. However, this effect was attenuated in tumors of endothelial-specific Trp53-knockout mice. Herein, we report that compared to X-ray irradiation, neutron radiation therapy reduced collagen deposition and suppressed EndMT in tumors. In addition to the fewer fibrotic changes, more cluster of differentiation (CD8)-positive cytotoxic T cells were observed in neutron-irradiated regrowing tumors than in X-ray-irradiated tumors. Furthermore, lower programmed death-ligand 1 (PD-L1) expression was noted in the former. Endothelial-specific Trp53 deletion suppressed fibrotic changes within the tumor environment following both X-ray and neutron radiation therapy. In particular, the upregulation in PD-L1 expression after X-ray radiation therapy was significantly dampened. Our findings suggest that compared to low LET radiation therapy, high LET radiation therapy can efficiently suppress profibrotic changes and enhance the anti-tumor immune response, resulting in delayed tumor regrowth.
Collapse
|
74
|
Braun C, Weichhart T. mTOR-dependent immunometabolism as Achilles' heel of anticancer therapy. Eur J Immunol 2021; 51:3161-3175. [PMID: 34648202 DOI: 10.1002/eji.202149270] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/07/2021] [Accepted: 10/06/2021] [Indexed: 12/14/2022]
Abstract
Immune cells are important constituents of the tumor microenvironment and essential in eradicating tumor cells during conventional therapies or novel immunotherapies. The mechanistic target of rapamycin (mTOR) signaling pathway senses the intra- and extracellular nutrient status, growth factor supply, and cell stress-related changes to coordinate cellular metabolism and activation dictating effector and memory functions in mainly all hematopoietic immune cells. In addition, the mTOR complex 1 (mTORC1) and mTORC2 are frequently deregulated and become activated in cancer cells to drive cell transformation, survival, neovascularization, and invasion. In this review, we provide an overview of the influence of mTOR complexes on immune and cancer cell function and metabolism. We discuss how mTOR inhibitors aiming to target cancer cells will influence immunometabolic cell functions participating either in antitumor responses or favoring tumor cell progression in individual immune cells. We suggest immunometabolism as the weak spot of anticancer therapy and propose to evaluate patients according to their predominant immune cell subtype in the cancer tissue. Advances in metabolic drug development that hold promise for more effective treatments in different types of cancer will have to consider their effects on the immune system.
Collapse
Affiliation(s)
- Clarissa Braun
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria.,Clinical Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Thomas Weichhart
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
75
|
Liu J, Chen J, Liu H, Zhang K, Zeng Q, Yang S, Jiang Z, Zhang X, Chen T, Li D, Shan H. Bi/Se-Based Nanotherapeutics Sensitize CT Image-Guided Stereotactic Body Radiotherapy through Reprogramming the Microenvironment of Hepatocellular Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2021; 13:42473-42485. [PMID: 34474563 DOI: 10.1021/acsami.1c11763] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The particular characteristics of hypoxia, immune suppression in the tumor microenvironment, and the lack of accurate imaging guidance lead to the limited effects of stereotactic body radiotherapy (SBRT) in reducing the recurrence rate and mortality of hepatocellular carcinoma (HCC). This research developed a novel theranostic agent based on Bi/Se nanoparticles (NPs), synthesized by a simple reduction reaction method for in vivo CT image-guided SBRT sensitization in mice. After loading Lenvatinib (Len), the obtained Bi/Se-Len NPs had excellent performance in reversing hypoxia and the immune suppression status of HCC. In vivo CT imaging results uncovered that the radiotherapy (RT) area could be accurately labeled after the injection of Bi/Se-Len NPs. Under Len's unique and robust properties, in vivo treatment was then carried out upon injection of Bi/Se-Len NPs, achieving excellent RT sensitization effects in a mouse HCC model. Comprehensive tests and histological stains revealed that Bi/Se-Len NPs could reshape and normalize tumor blood vessels, reduce the hypoxic situation of the tumor, and upregulate tumor-infiltrating CD4+ and CD8+ T lymphocytes around the tumors. Our work highlights an excellent proposal of Bi/Se-Len NPs as theranostic nanoparticles for image-guided HCC radiotherapy.
Collapse
Affiliation(s)
- Jiani Liu
- Center for Interventional Medicine, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
- The Cancer Center of The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
| | - Jiayao Chen
- Center for Interventional Medicine, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
| | - Hongxing Liu
- Department of Chemistry, Jinan University, Guangzhou, Guangdong Province, 510632, P. R. China
| | - Ke Zhang
- Center for Interventional Medicine, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
| | - Qi Zeng
- The Cancer Center of The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
| | - Shuai Yang
- The Cancer Center of The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
| | - Zebo Jiang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
| | - Xiaoting Zhang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
| | - Tianfeng Chen
- Department of Chemistry, Jinan University, Guangzhou, Guangdong Province, 510632, P. R. China
| | - Dan Li
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
| | - Hong Shan
- Center for Interventional Medicine, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, P. R. China
| |
Collapse
|
76
|
Mu Q, Najafi M. Modulation of the tumor microenvironment (TME) by melatonin. Eur J Pharmacol 2021; 907:174365. [PMID: 34302814 DOI: 10.1016/j.ejphar.2021.174365] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/10/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022]
Abstract
The tumor microenvironment (TME) includes a number of non-cancerous cells that affect cancer cell survival. Although CD8+ T lymphocytes and natural killer (NK) cells suppress tumor growth through induction of cell death in cancer cells, there are various immunosuppressive cells such as regulatory T cells (Tregs), tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs), myeloid-derived suppressor cells (MDSCs), etc., which drive cancer cell proliferation. These cells may also support tumor growth and metastasis by stimulating angiogenesis, epithelial-mesenchymal transition (EMT), and resistance to apoptosis. Interactions between cancer cells and other cells, as well as molecules released into EMT, play a key role in tumor growth and suppression of antitumoral immunity. Melatonin is a natural hormone that may be found in certain foods and is also available as a drug. Melatonin has been demonstrated to modulate cell activity and the release of cytokines and growth factors in TME. The purpose of this review is to explain the cellular and molecular mechanisms of cancer cell resistance as a result of interactions with TME. Next, we explain how melatonin affects cells and interactions within the TME.
Collapse
Affiliation(s)
- Qi Mu
- College of Nursing, Inner Mongolia University for Nationalities, Tongliao, 028000, China.
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
77
|
Gu X, Shi Y, Dong M, Jiang L, Yang J, Liu Z. Exosomal transfer of tumor-associated macrophage-derived hsa_circ_0001610 reduces radiosensitivity in endometrial cancer. Cell Death Dis 2021; 12:818. [PMID: 34462422 PMCID: PMC8405633 DOI: 10.1038/s41419-021-04087-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 07/22/2021] [Accepted: 07/27/2021] [Indexed: 02/07/2023]
Abstract
The occurrence of radioresistance is a clinical obstacle to endometrial cancer (EC) treatment and induces tumor relapse. In this study, we found that tumor-associated macrophages (TAMs) enriched in EC specimens were determined to present an M2-like phenotype. In vitro, the coculture of M2-polarized macrophages significantly downregulated the radiosensitivity of EC cells by releasing exosomes. Hsa_circ_0001610 was found to be abundant in exosomes derived from M2-polarized macrophages (EXOs), and hsa_circ_0001610 knockdown eliminated the reduction effect of EXOs on the radiosensitivity of EC cells. The following mechanism research revealed that hsa_circ_0001610 functioned as the competing endogenous RNA of miR-139-5p, thereby upregulating cyclin B1 expression, which is a vital pusher of radioresistance in several types of cancer by regulating the cell cycle. Hsa_circ_0001610 overexpression reduced the radiosensitivity of EC cells, which was then reversed by miR-139-5p overexpression. In vivo, the promotion effect of EXOs on xenograft tumor growth in nude mice treated with irradiation was further reinforced after hsa_circ_0001610 overexpression. In conclusion, TAM-derived exosomes transferred hsa_circ_0001610 to EC cells, and the overexpressed hsa_circ_0001610 in EC cells released cyclin B1 expression through adsorbing miR-139-5p, thereby weakening the radiosensitivity of EC cells.
Collapse
Affiliation(s)
- Xiaobin Gu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Rd, Zhengzhou, 450000, Henan, People's Republic of China.
| | - Yonggang Shi
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Rd, Zhengzhou, 450000, Henan, People's Republic of China
| | - Meilian Dong
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Rd, Zhengzhou, 450000, Henan, People's Republic of China
| | - Li Jiang
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Rd, Zhengzhou, 450000, Henan, People's Republic of China
| | - Jing Yang
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Rd, Zhengzhou, 450000, Henan, People's Republic of China
| | - Zheyan Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Rd, Zhengzhou, 450000, Henan, People's Republic of China
| |
Collapse
|
78
|
Du Z, Cai S, Yan D, Li H, Zhang X, Yang W, Cao J, Yi N, Tang Z. Development and Validation of a Radiosensitivity Prediction Model for Lower Grade Glioma Based on Spike-and-Slab Lasso. Front Oncol 2021; 11:701500. [PMID: 34395274 PMCID: PMC8363254 DOI: 10.3389/fonc.2021.701500] [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: 04/28/2021] [Accepted: 07/16/2021] [Indexed: 12/25/2022] Open
Abstract
Background and Purpose Lower grade glioma (LGG) is one of the leading causes of death world worldwide. We attempted to develop and validate a radiosensitivity model for predicting the survival of lower grade glioma by using spike-and-slab lasso Cox model. Methods In this research, differentially expressed genes based on tumor microenvironment was obtained to further analysis. Log-rank test was used to identify genes in patients who received radiotherapy and patients who did not receive radiotherapy, respectively. Then, spike-and-slab lasso was performed to select genes in patients who received radiotherapy. Finally, three genes (INA, LEPREL1 and PTCRA) were included in the model. A radiosensitivity-related risk score model was established based on overall rate of TCGA dataset in patients who received radiotherapy. The model was validated in TCGA dataset that PFS as endpoint and two CGGA datasets that OS as endpoint. A novel nomogram integrated risk score with age and tumor grade was developed to predict the OS of LGG patients. Results We developed and verified a radiosensitivity-related risk score model. The radiosensitivity-related risk score is served as an independent prognostic indicator. This radiosensitivity-related risk score model has prognostic prediction ability. Moreover, the nomogram integrated risk score with age and tumor grade was established to perform better for predicting 1, 3, 5-year survival rate. Conclusions This model can be used by clinicians and researchers to predict patient’s survival rates and achieve personalized treatment of LGG.
Collapse
Affiliation(s)
- Zixuan Du
- Department of Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Shang Cai
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Derui Yan
- Department of Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Huijun Li
- Department of Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Xinyan Zhang
- School of Data Science and Analytics, Kennesaw State University, Kennesaw, GA, United States
| | - Wei Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Jianping Cao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Nengjun Yi
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Zaixiang Tang
- Department of Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
79
|
Lai JZ, Zhu YY, Liu Y, Zhou LL, Hu L, Chen L, Zhang QY. Abscopal Effects of Local Radiotherapy Are Dependent on Tumor Immunogenicity. Front Oncol 2021; 11:690188. [PMID: 34249740 PMCID: PMC8264447 DOI: 10.3389/fonc.2021.690188] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 06/02/2021] [Indexed: 01/10/2023] Open
Abstract
Although abscopal tumor regression remains a rare phenomenon, interest in exploiting how radiation stimulates the immune system to induce systemic abscopal response is increasing. Here, we tested the hypothesis that tumor immunogenicity determined the ability of radiotherapy to induce abscopal effects. We established highly (MC-38 and E.G7-OVA) or poorly (LL/2 and B16-F10) immunogenic tumor models in this study and treated them with sham radiation, a single dose of 15 Gy, or three fractions of 5 Gy on three consecutive days. Alterations in the tumor microenvironment after radiation were examined by flow cytometry and RNA sequencing. Our results demonstrated the positive correlation between tumor immunogenicity and the abscopal effect of radiotherapy. The single dose of 15 Gy radiation was an effective regimen for inducing abscopal effects in highly immunogenic tumors. Local radiation reshaped the tumor microenvironment of irradiated and non-irradiated distant tumors by increasing CD8 T-cell infiltration and reducing suppressive immune cell accumulation. However, radiation alone was insufficient to elicit abscopal effects in poorly immunogenic tumors. No significant alterations were detected in the non-irradiated distant tumor microenvironment after radiation of poorly immunogenic tumors. In addition, tumor immunogenic subtypes were associated with the radiological response and clinical outcome of patients receiving radiotherapy. These findings indicated that tumor immunogenicity was the dominant characteristic that could predict the abscopal effect of radiotherapy. Our study provides an in-depth understanding of the immunological mechanisms involved in abscopal effects and highlights the impact of tumor heterogeneity on the therapeutic efficacy of radiotherapy and their combination with immunotherapy in clinical trials.
Collapse
Affiliation(s)
- Jin-Zhi Lai
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China.,Department of Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yan-Yang Zhu
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| | - Ying Liu
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| | - Lin-Lin Zhou
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| | - Li Hu
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| | - Ling Chen
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| | - Qiu-Yu Zhang
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| |
Collapse
|
80
|
Wang Q, Li S, Qiao S, Zheng Z, Duan X, Zhu X. Changes in T Lymphocyte Subsets in Different Tumors Before and After Radiotherapy: A Meta-analysis. Front Immunol 2021; 12:648652. [PMID: 34220806 PMCID: PMC8242248 DOI: 10.3389/fimmu.2021.648652] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 06/01/2021] [Indexed: 01/10/2023] Open
Abstract
Purpose Radiation therapy (RT) induces an immune response, but the relationship of this response with tumor type is not fully understood. This meta-analysis further elucidated this relationship by analyzing the changes in T lymphocyte subsets in different tumors before and after radiotherapy. Methods We searched English-language electronic databases including PubMed, EMBASE, and the Cochrane Library to collect studies on the changes in peripheral blood CD3+ T lymphocytes, CD4+ T lymphocytes, and CD8+ T lymphocytes before and after radiotherapy in tumor patients from January 2015 to April 2021. The quality of the included literature was evaluated using the NOS scale provided by the Cochrane Collaboration, and statistical software RevMan 5.4 was used to analyze the included literature. P<0.05 was considered to indicate statistical significance. Results A total of 19 studies in 16 articles involving 877 tumor patients were included. All data were collected within 1 month before or after radiotherapy. Meta-analysis showed that numbers of CD3+ T lymphocytes (SMD: -0.40; 95% CI [-0.75, -0.04]; p = 0.03) and CD4+ T lymphocytes (SMD: -0.43; 95% CI: [-0.85, -0.02]; p = 0.04) were significantly reduced after radiotherapy compared with before treatment, but there was no statistically significant difference for CD8+ T lymphocytes (SMD: 0.33; 95% CI: [-0.88, 0.74]; p = 0.12). Subgroup analysis showed that peripheral blood T lymphocytes decreased in head and neck cancer. However, in prostate cancer and breast cancer, there was no significant change in peripheral blood. 1 month after radiotherapy, it has a potential proliferation and activation effect on lymphocytes in esophageal cancer and lung cancer. The results showed that CD8+T lymphocytes increased in peripheral blood after SBRT. Radiotherapy alone reduced CD3+ T lymphocyte numbers. Conclusions Within 1 month of radiotherapy, patients have obvious immunological changes, which can cause apoptosis and reduction of T lymphocytes, and affect the balance of peripheral blood immune cells. The degree of immune response induced by radiotherapy differed between tumor types.
Collapse
Affiliation(s)
- Qin Wang
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shangbiao Li
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Simiao Qiao
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zhihao Zheng
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaotong Duan
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoxia Zhu
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
81
|
Nandi A, Chakrabarti R. The many facets of Notch signaling in breast cancer: toward overcoming therapeutic resistance. Genes Dev 2021; 34:1422-1438. [PMID: 33872192 PMCID: PMC7608750 DOI: 10.1101/gad.342287.120] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In this review, Nandi et al. revisit the mechanisms by which Notch receptors and ligands contribute to normal mammary gland development and breast tumor progression. The authors also discuss combinatorial approaches aimed at disrupting Notch- and TME-mediated resistance that may improve prognosis in breast cancer patients. Breast cancer is the second leading cause of cancer-related death in women and is a complex disease with high intratumoral and intertumoral heterogeneity. Such heterogeneity is a major driving force behind failure of current therapies and development of resistance. Due to the limitations of conventional therapies and inevitable emergence of acquired drug resistance (chemo and endocrine) as well as radio resistance, it is essential to design novel therapeutic strategies to improve the prognosis for breast cancer patients. Deregulated Notch signaling within the breast tumor and its tumor microenvironment (TME) is linked to poor clinical outcomes in treatment of resistant breast cancer. Notch receptors and ligands are also important for normal mammary development, suggesting the potential for conserved signaling pathways between normal mammary gland development and breast cancer. In this review, we focus on mechanisms by which Notch receptors and ligands contribute to normal mammary gland development and breast tumor progression. We also discuss how complex interactions between cancer cells and the TME may reduce treatment efficacy and ultimately lead to acquired drug or radio resistance. Potential combinatorial approaches aimed at disrupting Notch- and TME-mediated resistance that may aid in achieving in an improved patient prognosis are also highlighted.
Collapse
Affiliation(s)
- Ajeya Nandi
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Rumela Chakrabarti
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
82
|
Shaheer K, Lakshmanan MD. Effect of Piperine in Combination with Gamma Radiation on A549 Cells. JOURNAL OF HEALTH AND ALLIED SCIENCES NU 2021. [DOI: 10.1055/s-0040-1722808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Abstract
Background Lung cancer is a major constrain that increases mortality globally. Radiotherapy is one of the treatment modalities against lung cancer. A high dose of targeted radiation is required to achieve the treatment efficacy of cell killing. After radiotherapy, eventual tumor progression and therapy resistance are still a consequence of patient who undertakes nonsurgical radiation therapy. Piperine, a plant alkaloid, has been known to enhance the action of the anticancer drugs in various drug-resistant cancer cells. The aim of the current in vitro study was to study the effect of piperine on radiosensitizing property against A549 cells.
Methods In vitro radiosensitizing activity of piperine was elucidated on A549 cells using MTT (3-(4, 5-dimethylthiazol-2-yl)-25-diphenyltetrazolium bromide) assay. CompuSyn analysis was used to compute the combination index values to analyze the combinatory effect of piperine and radiation
Results and Conclusion We observed that piperine increased tumor cell killing in combination with the γ-radiation in vitro. However, further studies are warranted to understand the molecular mechanism of the radiosensitizing action of piperine.
Collapse
Affiliation(s)
- Koniyan Shaheer
- Molecular Biology Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka, India
| | - M. Divya Lakshmanan
- Molecular Biology Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka, India
| |
Collapse
|
83
|
Wang Y, Shen N, Wang Y, Li M, Zhang W, Fan L, Liu L, Tang Z, Chen X. Cisplatin nanoparticles boost abscopal effect of radiation plus anti-PD1 therapy. Biomater Sci 2021; 9:3019-3027. [PMID: 33656040 DOI: 10.1039/d1bm00112d] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The abscopal effect of radiation therapy (RT) is clinically significant but occurs rarely. Although anti-programmed cell death protein 1 antibody (anti-PD1) is likely to enhance the abscopal effect in patients receiving RT, the incidence rate remains less than 30%. One major limitation is the paucity of CD8+ T cells within non-irradiated tumors. Here, cisplatin (CDDP) loaded poly(l-glutamic acid)-graft-methoxy poly(ethylene glycol) complex nanoparticles (CDDP-NPs) are confirmed to increase CD8+ T cells within non-irradiated tumors and boost the abscopal effect of RT plus anti-PD1, and more strongly than CDDP. Compared to RT and RT + CDDP, RT + CDDP-NPs induced greater immunogenic cell death (ICD) with enhanced proportion of Calreticulin+ Lewis lung cancer (LLC) cells (16.47%, 20.53% and 27.03%), along with which more CD8+ T cells were infiltrated into CDDP-NP treated irradiated tumors in the unilateral LLC tumor model. In the bilateral LLC tumor model, RT + CDDP-NPs significantly induced more chemokine (C-X-C motif) ligand 10 (CXCL10) secretion (36.3, 44.19 and 56.37 pg mL-1), which corresponded to greater CD8+ T cell infiltration in the non-irradiated tumors (0.19%, 0.20% and 0.72%). Finally, compared to RT + anti-PD1 and RT + anti-PD1 + CDDP, RT + anti-PD1 + CDDP-NPs significantly inhibited the growth of non-irradiated tumors more forcefully, as indicated by the respective tumor volumes of 1141, 1146 and 585 mm3. This is the first study to show that CDDP-NPs can amplify RT-induced immune activation and break through the efficiency limitation of the RT plus anti-PD1 induced abscopal effect.
Collapse
Affiliation(s)
- Ying Wang
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun 130041, PR China.
| | - Na Shen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China.
| | - Yue Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China.
| | - Mo Li
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun 130041, PR China.
| | - Wanze Zhang
- Department of Pathogenobiology, College of Basic Medical Science, Jilin University, Changchun 130021, PR China
| | - Liwen Fan
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun 130041, PR China.
| | - Linlin Liu
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun 130041, PR China.
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China.
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China.
| |
Collapse
|
84
|
Hu S, Zhu L, Song Y, Zhao X, Chen Q, Pan Y, Zhang J, Bai Y, Zhang H, Shao C. Radiation-induced abscopal reproductive effect is driven by TNF-α/p38 MAPK/Rac1 axis in Sertoli cells. Am J Cancer Res 2021; 11:5742-5758. [PMID: 33897879 PMCID: PMC8058717 DOI: 10.7150/thno.56853] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 03/03/2021] [Indexed: 12/24/2022] Open
Abstract
Rationale: Radiotherapy has become a mainstay for tumor management, and more than 50% of patients with thoracic tumor need to be treated with radiotherapy. However, the potential adverse effects of thoracic radiotherapy on the reproductive system remain elusive. Methods: Western blot analysis, immunofluorescence assay and transmission electron microscopy (TEM) analysis were performed to investigate the integrity of blood-testis barrier (BTB) in male mice after hypofractionated irradiation (IR) on the right thorax. RNA sequencing, co-immunoprecipitation (IP), Duolink PLA and inhibitor experiments were carried out to demonstrate the molecular mechanisms of the BTB dynamics changes and the subsequent reproductive effect. Results: It was found that the hypofractionated IR on right thorax evoked ultrastructural destruction in distant testes, and thus caused radiation-induced abscopal reproductive effect (RIARE) in male mice. Mechanistically, thoracic IR induced significant nuclear translocation of Rac Family Small GTPase 1 (Rac1) in abscopal Sertoli cells, which closely correlated with the activation of TNF-α/p38 mitogen activated protein kinase (MAPK) pathway. Of note, YWHAZ, a critical polarity protein, was found to be co-localized with Rac1 in Sertoli cells, and this interaction was indispensable for thoracic IR-induced Rac1 nuclear translocation and subsequent degradation of BTB-associated proteins. Conclusions: Our findings imply for the first time that YWHAZ-mediated Rac1 nuclear translocation plays central roles in RIARE, and TNF-α/p38 MAPK/Rac1 axis can be employed as a therapeutic target against RIARE for young male patients receiving hypofractionated radiotherapy.
Collapse
|
85
|
Liu Q, Hao Y, Du R, Hu D, Xie J, Zhang J, Deng G, Liang N, Tian T, Käsmann L, Rades D, Rim CH, Hu P, Zhang J. Radiotherapy programs neutrophils to an antitumor phenotype by inducing mesenchymal-epithelial transition. Transl Lung Cancer Res 2021; 10:1424-1443. [PMID: 33889520 PMCID: PMC8044478 DOI: 10.21037/tlcr-21-152] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 03/25/2021] [Indexed: 11/06/2022]
Abstract
BACKGROUND Neutrophils can play a pro-tumor or anti-tumor role depending on the tumor microenvironment. The effects of concurrent treatment with granulocyte colony-stimulating factor (G-CSF) and radiotherapy (RT) on neutrophils have not yet to be described. METHODS Hypofractionated radiation of 8 Gy ×3 fractions was administered with or without recombinant G-CSF to Lewis lung carcinoma tumor-bearing C57BL/6 model mice. The activation status of cytotoxic T cells in the mice was measured, along with the levels of tumor-associated neutrophils, cytotoxic T cells, and Treg cells. Tumor growth, survival, cytokine expression, and signaling pathways underlying anti-tumor effects of tumor-associated neutrophils after treatment were also studied. To ascertain the effects of concurrent RT and G-CSF on tumor-associated neutrophils, neutrophil depletion was performed. RESULTS RT affected early neutrophil infiltration, which is the first-line immune response. Subsequently, enhanced accumulation of lymphocytes, particularly CD8 cytotoxic T cells, was observed. Notably, lymphocytic infiltration was inhibited by neutrophil depletion but enhanced by G-CSF treatment. RT generated persistent DNA damage, as evidenced by an accumulation of phosphorylation of histone H2AX (γH2AX), and subsequently triggered inflammatory chemokine secretion. The chemokines CXCL1, CXCL2, and CCL5 were upregulated in both radiation-treated cells and the corresponding supernatants. Neutrophils that were newly recruited after RT improved radiosensitivity by inhibiting epithelial-mesenchymal transition via the reactive oxygen species-mediated PI3K/Akt/Snail signaling pathway, and G-CSF treatment enhanced this effect. CONCLUSIONS The results of this study suggest that RT activates neutrophil recruitment and polarizes newly recruited neutrophils toward an antitumor phenotype, which is enhanced by the concurrent administration of G-CSF. Mesenchymal-epithelial transition induced by reactive oxygen species accumulation plays a major role in this process. Thus, the polarization of tumor-associated neutrophils might play a role in future cancer immunotherapies.
Collapse
Affiliation(s)
- Qiqi Liu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shandong Lung Cancer Institute, Jinan, China
| | - Yuying Hao
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
- Department of Radiation Oncology, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Rui Du
- Division of Oncology, Department of Graduate, Weifang Medical College, Weifang, China
| | - Dan Hu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Department of Physiology, Jeonbuk National University Medical School, Jeonju 54907, Jeollabuk-do, Korea
| | - Jian Xie
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
| | - Jingxin Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China
| | - Guodong Deng
- Department of Chemical Etiology and Carcinogenesis, Cancer Institute, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Ning Liang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
| | - Tiantian Tian
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
| | - Lukas Käsmann
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Dirk Rades
- Department of Radiation Oncology, University of Lübeck, Lübeck, Germany
| | - Chai Hong Rim
- Department of Radiation Oncology, Korea University Ansan Hospital, Ansan, Republic of Korea
| | - Pingping Hu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
| | - Jiandong Zhang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
| |
Collapse
|
86
|
Li Z, Li Y, Gao J, Fu Y, Hua P, Jing Y, Cai M, Wang H, Tong T. The role of CD47-SIRPα immune checkpoint in tumor immune evasion and innate immunotherapy. Life Sci 2021; 273:119150. [PMID: 33662426 DOI: 10.1016/j.lfs.2021.119150] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/20/2021] [Accepted: 01/26/2021] [Indexed: 02/07/2023]
Abstract
As a transmembrane protein, CD47 plays an important role in mediating cell proliferation, migration, phagocytosis, apoptosis, immune homeostasis, inhibition of NO signal transduction and other related reactions. Upon the interaction of innate immune checkpoint CD47-SIRPα occurrence, they send a "don't eat me" signal to the macrophages. This signal ultimately helps tumors achieve immune escape by inhibiting macrophage contraction to prevent tumor cells from phagocytosis. Therefore, the importance of CD47-SIRPα immune checkpoint inhibitors in tumor immunotherapy has attracted more attention in recent years. Based on the cognitive improvement of the effect with CD47 in tumor microenvironment and tumor characteristics, the pace of tumor treatment strategies for CD47-SIRPα immune checkpoint inhibitors has gradually accelerated. In this review, we introduced the high expression of CD47 in cancer cells to avoid phagocytosis by immune cells and the importance of CD47 in the structure of cancer microenvironment and the maintenance of cancer cell characteristics. Given the role of the innate immune system in tumorigenesis and development, an improved understanding of the anti-tumor process of innate immune checkpoint inhibitors can lay the foundation for more effective combinations with other anti-tumor treatment strategies.
Collapse
Affiliation(s)
- Zihao Li
- The Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Yue Li
- The Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Jing Gao
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Yilin Fu
- The Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Peiyan Hua
- The Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Yingying Jing
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China; University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Mingjun Cai
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Hongda Wang
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China; University of Science and Technology of China, Hefei, Anhui 230027, China; Laboratory for Marine Biology and Biotechnology, Qing dao National Laboratory for Marine Science and Technology, Wenhai Road, Aoshanwei, Jimo, Qingdao, Shandong 266237, China
| | - Ti Tong
- The Second Hospital of Jilin University, Changchun, Jilin 130041, China.
| |
Collapse
|
87
|
Tubin S, Khan MK, Gupta S, Jeremic B. Biology of NSCLC: Interplay between Cancer Cells, Radiation and Tumor Immune Microenvironment. Cancers (Basel) 2021; 13:775. [PMID: 33673332 PMCID: PMC7918834 DOI: 10.3390/cancers13040775] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/12/2022] Open
Abstract
The overall prognosis and survival of non-small cell lung cancer (NSCLC) patients remain poor. The immune system plays an integral role in driving tumor control, tumor progression, and overall survival of NSCLC patients. While the tumor cells possess many ways to escape the immune system, conventional radiotherapy (RT) approaches, which are directly cytotoxic to tumors, can further add additional immune suppression to the tumor microenvironment by destroying many of the lymphocytes that circulate within the irradiated tumor environment. Thus, the current immunogenic balance, determined by the tumor- and radiation-inhibitory effects is significantly shifted towards immunosuppression, leading to poor clinical outcomes. However, newer emerging evidence suggests that tumor immunosuppression is an "elastic process" that can be manipulated and converted back into an immunostimulant environment that can actually improve patient outcome. In this review we will discuss the natural immunosuppressive effects of NSCLC cells and conventional RT approaches, and then shift the focus on immunomodulation through novel, emerging immuno- and RT approaches that promise to generate immunostimulatory effects to enhance tumor control and patient outcome. We further describe some of the mechanisms by which these newer approaches are thought to be working and set the stage for future trials and additional preclinical work.
Collapse
Affiliation(s)
- Slavisa Tubin
- MedAustron Ion Therapy Center, Marie Curie-Straße 5, 2700 Wiener Neustadt, Austria
| | - Mohammad K. Khan
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, 1365-C Clifton Road, Atlanta, GA 30322, USA;
| | - Seema Gupta
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA;
| | - Branislav Jeremic
- Research Institute of Clinical Medicine, 13 Tevdore Mgdveli, Tbilisi 0112, Georgia;
| |
Collapse
|
88
|
Ren J, Xu M, Chen J, Ding J, Wang P, Huo L, Li F, Liu Z. PET imaging facilitates antibody screening for synergistic radioimmunotherapy with a 177Lu-labeled αPD-L1 antibody. Theranostics 2021; 11:304-315. [PMID: 33391476 PMCID: PMC7681088 DOI: 10.7150/thno.45540] [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: 03/02/2020] [Accepted: 09/21/2020] [Indexed: 12/11/2022] Open
Abstract
Rationale: The low response rate of immunotherapy, such as anti-PD-L1/PD-1 and anti-CTLA4, has limited its application to a wider population of cancer patients. One widely accepted view is that inflammation within the tumor microenvironment is low or ineffective for inducing the sufficient infiltration and/or activation of lymphocytes. Here, a highly tumor-selective anti-PD-L1 (αPD-L1) antibody was developed through PET imaging screening, and it was radiolabeled with Lu-177 for PD-L1-targeted radioimmunotherapy (RIT) and radiation-synergized immunotherapy. Methods: A series of αPD-L1 antibodies were radiolabeled with zirconium-89 for PET imaging to screen the most suitable antibodies for RIT. Mice were divided into an immunotherapy group, a RIT group and a radiation-synergized immunotherapy group to evaluate the therapeutic effect. Alterations in the tumor microenvironment after treatment were assessed using flow cytometry and immunofluorescence microscopy. Results: Radiation-synergistic RIT can achieve a significantly better therapeutic effect than immunotherapy or RIT alone. The dosages of the radiopharmaceuticals and αPD-L1 antibodies were reduced, the infiltration of CD4+ and CD8+ T cells in the tumor microenvironment was increased, and no side effects were observed. This radiation-synergistic RIT strategy successfully showed a strong synergistic effect with αPD-L1 checkpoint blockade therapy, at least in the mouse model. Conclusions: PET imaging of 89Zr-labeled antibodies is an effective method for antibody screening. RIT with a 177Lu-labeled αPD-L1 antibody could successfully upregulate antitumor immunity in the tumor microenvironment and turn "cold" tumors "hot" for immunotherapy.
Collapse
|
89
|
Shah A, Rauth S, Aithal A, Kaur S, Ganguly K, Orzechowski C, Varshney GC, Jain M, Batra SK. The Current Landscape of Antibody-based Therapies in Solid Malignancies. Am J Cancer Res 2021; 11:1493-1512. [PMID: 33391547 PMCID: PMC7738893 DOI: 10.7150/thno.52614] [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: 08/30/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023] Open
Abstract
Over the past three decades, monoclonal antibodies (mAbs) have revolutionized the landscape of cancer therapy. Still, this benefit remains restricted to a small proportion of patients due to moderate response rates and resistance emergence. The field has started to embrace better mAb-based formats with advancements in molecular and protein engineering technologies. The development of a therapeutic mAb with long-lasting clinical impact demands a prodigious understanding of target antigen, effective mechanism of action, gene engineering technologies, complex interplay between tumor and host immune system, and biomarkers for prediction of clinical response. This review discusses the various approaches used by mAbs for tumor targeting and mechanisms of therapeutic resistance that is not only caused by the heterogeneity of tumor antigen, but also the resistance imposed by tumor microenvironment (TME), including inefficient delivery to the tumor, alteration of effector functions in the TME, and Fc-gamma receptor expression diversity and polymorphism. Further, this article provides a perspective on potential strategies to overcome these barriers and how diagnostic and prognostic biomarkers are being used in predicting response to mAb-based therapies. Overall, understanding these interdependent parameters can improve the current mAb-based formulations and develop novel mAb-based therapeutics for achieving durable clinical outcomes in a large subset of patients.
Collapse
|
90
|
Yang Q, Guo N, Zhou Y, Chen J, Wei Q, Han M. The role of tumor-associated macrophages (TAMs) in tumor progression and relevant advance in targeted therapy. Acta Pharm Sin B 2020; 10:2156-2170. [PMID: 33304783 PMCID: PMC7714989 DOI: 10.1016/j.apsb.2020.04.004] [Citation(s) in RCA: 246] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 03/24/2020] [Accepted: 03/27/2020] [Indexed: 12/17/2022] Open
Abstract
Macrophages have a leading position in the tumor microenvironment (TME) which paves the way to carcinogenesis. Initially, monocytes and macrophages are recruited to the sites where the tumor develops. Under the guidance of different microenvironmental signals, macrophages would polarize into two functional phenotypes, named as classically activated macrophages (M1) and alternatively activated macrophages (M2). Contrary to the anti-tumor effect of M1, M2 exerts anti-inflammatory and tumorigenic characters. In progressive tumor, M2 tumor-associated macrophages (TAMs) are in the majority, being vital regulators reacting upon TME. This review elaborates on the role of TAMs in tumor progression. Furthermore, prospective macrophage-focused therapeutic strategies, including drugs not only in clinical trials but also at primary research stages, are summarized followed by a discussion about their clinical application values. Nanoparticulate systems with efficient drug delivery and improved antitumor effect are also summed up in this article.
Collapse
Affiliation(s)
- Qiyao Yang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ningning Guo
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yi Zhou
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiejian Chen
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Qichun Wei
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Min Han
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
91
|
Hu Z, Cai B, Wang M, Wen X, Geng A, Hu X, Xue R, Mao Z, Jiang Y, Wan X. Diosmetin enhances the sensitivity of radiotherapy by suppressing homologous recombination in endometrial cancer. Cell Cycle 2020; 19:3115-3126. [PMID: 33064975 DOI: 10.1080/15384101.2020.1831257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Radiotherapy is an essential treatment for endometrial cancer (EC), especially in advanced, metastatic, and recurrent cases. Combining radiotherapy, which mainly causes DNA double-strand breaks (DSBs), with small molecules targeting aberrantly activated homologous recombination (HR) repair pathways holds great potential for treating ECs in advanced stages. Here, we demonstrate that diosmetin (DIO), a natural flavonoid, suppresses HR, therefore inhibiting cell proliferation and enhancing the sensitivity of EC to radiotherapy. Clonogenic experiments revealed that combining DIO and X-ray significantly inhibited the viability of EC cells compared to cells treated with diosmetin or X-ray alone. The survival fraction of EC cells decreased to 40% when combining 0.4 Gy X-ray and 4 μM DIO; however, each treatment alone only caused death in approximately 15% and 22% of cancer cells, respectively. Further mechanistic studies showed that diosmetin inhibited the recruitment of RPA2 and RAD51, two critical factors involved in the HR repair pathway, upon the occurrence of DSBs. Thus, we propose that a combination of diosmetin and irradiation is a promising therapeutic strategy for treating endometrial cancer.
Collapse
Affiliation(s)
- Zhiyi Hu
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, School of Medicine, Tongji University , Shanghai, China
| | - Bailian Cai
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, School of Medicine, Tongji University , Shanghai, China
| | - Mengfei Wang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, School of Medicine, Tongji University , Shanghai, China
| | - Xiaoli Wen
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, School of Medicine, Tongji University , Shanghai, China
| | - Anke Geng
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, School of Medicine, Tongji University , Shanghai, China.,Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University , Shanghai, China
| | - Xiang Hu
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, School of Medicine, Tongji University , Shanghai, China
| | - Renhao Xue
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, School of Medicine, Tongji University , Shanghai, China
| | - Zhiyong Mao
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, School of Medicine, Tongji University , Shanghai, China.,Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University , Shanghai, China
| | - Ying Jiang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University , Shanghai, China
| | - Xiaoping Wan
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, School of Medicine, Tongji University , Shanghai, China.,Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University , Shanghai, China
| |
Collapse
|
92
|
Radiotherapy-Mediated Immunomodulation and Anti-Tumor Abscopal Effect Combining Immune Checkpoint Blockade. Cancers (Basel) 2020; 12:cancers12102762. [PMID: 32992835 PMCID: PMC7600068 DOI: 10.3390/cancers12102762] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/19/2020] [Accepted: 09/23/2020] [Indexed: 12/17/2022] Open
Abstract
Radiotherapy (RT) is a conventional method for clinical treatment of local tumors, which can induce tumor-specific immune response and cause the shrinkage of primary tumor and distal metastases via mediating tumor infiltration of CD8+ T cells. Ionizing radiation (IR) induced tumor regression outside the radiation field is termed as abscopal effect. However, due to the mobilization of immunosuppressive signals by IR, the activated CD8+T cells are not sufficient to maintain a long-term positive feedback to make the tumors regress completely. Eventually, the "hot" tumors gradually turn to "cold". With the advent of emerging immunotherapy, the combination of immune checkpoint blockade (ICB) and local RT has produced welcome changes in stubborn metastases, especially anti-PD-1/PD-L1 and anti-CTLA-4 which have been approved in clinical cancer treatment. However, the detailed mechanism of the abscopal effect induced by combined therapy is still unclear. Therefore, how to formulate a therapeutic schedule to maximize the efficacy should be took into consideration according to specific circumstance. This paper reviewed the recent research progresses in immunomodulatory effects of local radiotherapy on the tumor microenvironment, as well as the unique advantage for abscopal effect when combined with ICB, with a view to exploring the potential application value of radioimmunotherapy in clinic.
Collapse
|
93
|
Kerr MD, McBride DA, Chumber AK, Shah NJ. Combining therapeutic vaccines with chemo- and immunotherapies in the treatment of cancer. Expert Opin Drug Discov 2020; 16:89-99. [PMID: 32867561 DOI: 10.1080/17460441.2020.1811673] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Breakthroughs in cancer immunotherapy have spurred interest in the development of vaccines to mediate prophylactic protection and therapeutic efficacy against primary tumors or to prevent relapse. However, immunosuppressive mechanisms employed by cancer cells to generate effective resistance have hampered clinical translation of therapeutic cancer vaccines. To enhance vaccine efficacy, the immunomodulatory properties of cytoreductive therapies could amplify a cancer-specific immune response. AREAS COVERED Herein, the authors discuss therapeutic cancer vaccines that harness whole cells and antigen-targeted vaccines. First, recent advancements in both autologous and allogeneic whole-cell vaccines and combinations with checkpoint blockade and chemotherapy are reviewed. Next, tumor antigen-targeted vaccines using peptide-based vaccines and DNA-vaccines are discussed. Finally, combination therapies using antigen-targeted vaccines are reviewed. EXPERT OPINION A deeper understanding of the immunostimulatory properties of cytoreductive therapies has supported their utility in combination therapies involving cancer vaccines as a potential strategy to induce a durable anti-tumor immune response for multiple types of cancers. Based on current evidence, combination therapies may have synergies that depend on the identity of the cytotoxic agent, vaccine target, dosing schedule, and cancer type. Together, these observations suggest that combining cancer vaccines with immunomodulatory cytoreductive therapy is a promising strategy for cancer therapy.
Collapse
Affiliation(s)
- Matthew D Kerr
- Department of NanoEngineering, University of California , San Diego, CA, USA.,Program in Chemical Engineering, University of California , San Diego, CA, USA.,Center for Nano-Immuno Engineering, University of California , San Diego, CA, USA
| | - David A McBride
- Department of NanoEngineering, University of California , San Diego, CA, USA.,Program in Chemical Engineering, University of California , San Diego, CA, USA.,Center for Nano-Immuno Engineering, University of California , San Diego, CA, USA
| | - Arun K Chumber
- Department of NanoEngineering, University of California , San Diego, CA, USA.,Program in Chemical Engineering, University of California , San Diego, CA, USA.,Center for Nano-Immuno Engineering, University of California , San Diego, CA, USA
| | - Nisarg J Shah
- Department of NanoEngineering, University of California , San Diego, CA, USA.,Program in Chemical Engineering, University of California , San Diego, CA, USA.,Center for Nano-Immuno Engineering, University of California , San Diego, CA, USA.,Program in Immunology, University of California , San Diego, CA, USA.,San Diego Center for Precision Immunotherapy, Moores Cancer Center, University of California , San Diego, CA, USA
| |
Collapse
|
94
|
Liu Z, Wang J, Liu L, Yuan H, Bu Y, Feng J, Liu Y, Yang G, Zhao M, Yuan Y, Zhang H, Yun H, Zhang X. Chronic ethanol consumption and HBV induce abnormal lipid metabolism through HBx/SWELL1/arachidonic acid signaling and activate Tregs in HBV-Tg mice. Am J Cancer Res 2020; 10:9249-9267. [PMID: 32802190 PMCID: PMC7415795 DOI: 10.7150/thno.46005] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 07/09/2020] [Indexed: 02/07/2023] Open
Abstract
Rationale: Chronic ethanol consumption as a public health problem worldwide boosts the development of chronic liver diseases in hepatitis B virus (HBV)-infected patients. Arachidonic acid metabolite prostaglandin E2 (PGE2) activates regulatory T cells (Tregs) function. Here, we aim to investigate the underlying mechanism by which chronic ethanol consumption enriches the HBV-induced abnormal lipid metabolism and Tregs. Methods: The si-RNAs were used to weaken the expression of SWELL1 in HepG2, HepG2.2.15 and K180 cancer cell lines, followed by RNA sequencing from HepG2 cells. Arachidonic acid metabolite PGE2 and LTD4 were measured by ELISA assay in vivo and in vitro. Western blot analysis and RT-qPCR were used to examine HBx and SWELL1 and transcriptional factor Sp1 in clinical HCC samples and cell lines. The effect of chronic ethanol consumption on Tregs was tested by flow cytometry in HBV-Tg mice. The splenic Tregs were collected and analyzed by RNA sequencing. Results: The cooperative effect of ethanol and HBV in abnormal lipid metabolism was observed in vivo and in vitro. The depression of SWELL1 (or HBx) resulted in the reduction of lipid content and arachidonic acid metabolite, correlating with suppression of relative gene atlas. Ethanol and SWELL1 elevated the levels of PGE2 or LTD4 in the liver of mice and cell lines. Interestingly, the ethanol modulated abnormal lipid metabolism through activating HBx/Sp1/SWELL1/arachidonic acid signaling. Chronic ethanol consumption remarkably increased the population of PBL Tregs and splenic Tregs in HBV-Tg mice, consistently with the enhanced expression of PD-L1 in vivo and in vitro. Mechanically, RNA-seq data showed that multiple genes were altered in the transcriptomic atlas of Tregs sorting from ethanol-fed mice or HBV-Tg mice. Conclusion: The chronic ethanol intake enriches the HBV-enhanced abnormal lipid metabolism through HBx/SWELL1/arachidonic acid signaling and activates Tregs in mice.
Collapse
|
95
|
Ashrafizadeh M, Farhood B, Eleojo Musa A, Taeb S, Najafi M. The interactions and communications in tumor resistance to radiotherapy: Therapy perspectives. Int Immunopharmacol 2020; 87:106807. [PMID: 32683299 DOI: 10.1016/j.intimp.2020.106807] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 06/26/2020] [Accepted: 07/10/2020] [Indexed: 12/24/2022]
Abstract
Tumor microenvironment (TME) includes a wide range of cell types including cancer cells, cells which are involved in stromal structure and immune cells (tumor suppressor and tumor promoting cells). These cells have several interactions with each other that are mainly regulated via the release of intercellular mediators. Radiotherapy can modulate these interactions via shifting secretions into inflammatory or anti-inflammatory responses. Radiotherapy also can trigger resistance of cancer (stem) cells via activation of stromal cells. The main mechanisms of tumor resistance to radiotherapy is the exhaustion of anti-tumor immunity via suppression of CD4+ T cells and apoptosis of cytotoxic CD8+ T lymphocytes (CTLs). Cancer-associated fibroblasts (CAFs), mesenchymal-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) are the main suppressor of anti-tumor immunity via the release of several chemokines, cytokines and immune suppressors. In this review, we explain the main cellular and molecular interactions and secretions in TME following radiotherapy. Furthermore, the main signaling pathways and intercellular connections that can be targeted to improve therapeutic efficiency of radiotherapy will be discussed.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Ahmed Eleojo Musa
- Department of Medical Physics, Tehran University of Medical Sciences (International Campus), Tehran, Iran
| | - Shahram Taeb
- Ionizing and Non-Ionizing Radiation Protection Research Center (INIRPRC), Shiraz University of Medical Sciences, Shiraz, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
96
|
Liao Y, Liu S, Fu S, Wu J. HMGB1 in Radiotherapy: A Two Headed Signal Regulating Tumor Radiosensitivity and Immunity. Onco Targets Ther 2020; 13:6859-6871. [PMID: 32764978 PMCID: PMC7369309 DOI: 10.2147/ott.s253772] [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: 03/13/2020] [Accepted: 06/19/2020] [Indexed: 12/14/2022] Open
Abstract
Radiotherapy (RT) is a mainstay of cancer treatment. Recent studies have shown that RT not only directly induces cell death but also has late and sustained immune effects. High mobility group box 1 (HMGB1) is a nuclear protein released during RT, with location-dependent functions. It is essential for normal cellular function but also regulates the proliferation and migration of tumor cells by binding to high-affinity receptors. In this review, we summarize recent evidence on the functions of HMGB1 in RT according to the position, intracellular HMGB1 and extracellular HMGB1. Intracellular HMGB1 induces radiation tolerance in tumor cells by promoting DNA damage repair and autophagy. Extracellular HMGB1 plays a more intricate role in radiation-related immune responses, wherein it not only stimulates the anti-tumor immune response by facilitating the recognition of dying tumor cells but is also involved in maintaining immunosuppression. Factors that potentially affect the role of HMGB1 in RT-induced cytotoxicity have also been discussed in the context of possible therapeutic applications, which helps to develop effective and targeted radio-sensitization therapies.
Collapse
Affiliation(s)
- Yin Liao
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Shuya Liu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Shaozhi Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Jingbo Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| |
Collapse
|
97
|
Ashrafizadeh M, Farhood B, Eleojo Musa A, Taeb S, Najafi M. Damage-associated molecular patterns in tumor radiotherapy. Int Immunopharmacol 2020; 86:106761. [PMID: 32629409 DOI: 10.1016/j.intimp.2020.106761] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/26/2020] [Accepted: 06/27/2020] [Indexed: 02/06/2023]
Abstract
Radiotherapy is one of the most common modalities for the treatment of cancer. One of the most promising effects of radiotherapy is immunologic cell death and the release of danger alarms, which are known as damage-associated molecular patterns (DAMPs). DAMPs are able to trigger cancer cells and other cells within tumor microenvironment (TME), either for suppression or promotion of tumor growth. Heat shock proteins (HSPs) including HSP70 and HSP90, high mobility group box 1 (HMGB1), and adenosine triphosphate (ATP) and its metabolites such as adenosine are the most common danger alarms that are released after radiotherapy-induced immunologic cell death. Some DAMPs including adenosine is able to interact with both cancer cells as well as other cells in TME to promote tumor growth and resistance to radiotherapy. However, others are able to trigger anti-tumor immunity or both tumor suppressive and immunosuppressive mechanisms depending on affected cells. In this review, we explain the mechanisms behind the release of radiation-induced DAMPs, and its consequences on cells within tumor. Targeting of these mechanisms may be in favor of tumor control in combination with radiotherapy and radioimmunotherapy.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Ahmed Eleojo Musa
- Department of Medical Physics, Tehran University of Medical Sciences (International Campus), Tehran, Iran
| | - Shahram Taeb
- Ionizing and Non-Ionizing Radiation Protection Research Center (INIRPRC), Shiraz University of Medical Sciences, Shiraz, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
98
|
Liu Q, Zhang D, Qian H, Chu Y, Yang Y, Shao J, Xu Q, Liu B. Superior Antitumor Efficacy of IFN-α2b-Incorporated Photo-Cross-Linked Hydrogels Combined with T Cell Transfer and Low-Dose Irradiation Against Gastric Cancer. Int J Nanomedicine 2020; 15:3669-3680. [PMID: 32547021 PMCID: PMC7261665 DOI: 10.2147/ijn.s249174] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/07/2020] [Indexed: 12/18/2022] Open
Abstract
Introduction The exhaustion and poor homing of activated lymphocytes are critical obstacles in adoptive cell immunotherapy for solid tumors. In order to effectively deliver immune cells into tumors, we encapsulated interferon-α2b (IFN-α2b) into macroporous hydrogels as an enhancement factor and utilized low-dose irradiation (LDI) as a tumoral attractor of T cells. Methods Hydroxypropyl cellulose hydrogels were prepared by irradiation techniques, and the cross-sectional microstructure was characterized by scanning electron microscopy. The synergistic antitumor mechanism of combination of IFN-α2b and CIK cells was evaluated by detecting the expression of activation marker CD69 on CIK cell surface and IFN-γ production by CIK cells. The in vivo antitumor activity of IFN-α2b-incorporated hydroxypropyl cellulose hydrogels combined with CIK and radiation was evaluated in an MKN-45 xenografted nude mice model. Results The bioactivity of IFN-α2b was well maintained in ultraviolet-reactive, rapidly cross-linkable hydroxypropyl cellulose hydrogels. In vitro studies demonstrated IFN-α2b-activated T cells, as evidenced by upregulating early activation marker CD69 and secretion inflammatory cytokine IFN-γ. In vivo real-time image showed our hydrogels kept a higher amount of drug delivery at the tumor site for a long time compared with free drug injection. Low-dose irradiation promoted T cell accumulation and infiltration in subcutaneous tumors. Combination of IFN-α2b-loaded hydrogels (Gel-IFN) with T cells and LDI exhibited higher efficacy to eradicate human gastric cancer xenograted tumors with less proliferating cells and more necrotic regions compared with IFN-α2b or T cells alone. Discussion HPC hydrogels kept the activity of IFN-α2b and stably release of IFN-α2b to stimulate T cells for a long time. At the same time, low-dose radiation recruits T cells into tumors. This innovative integration mode of IFN-α2b-loaded hydrogels and radiotherapy offers a potent strategy to improve the therapeutic outcome of T cell therapy.
Collapse
Affiliation(s)
- Qin Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China
| | - Dinghu Zhang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China.,Department of Radiology, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China
| | - Hanqing Qian
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China
| | - Yanhong Chu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China
| | - Yan Yang
- Department of Oncology, Jiangning Hospital, Nanjing, People's Republic of China
| | - Jie Shao
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China
| | - Qiuping Xu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China
| | - Baorui Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, People's Republic of China
| |
Collapse
|
99
|
Chen X, Yang J, Wang L, Liu B. Personalized neoantigen vaccination with synthetic long peptides: recent advances and future perspectives. Theranostics 2020; 10:6011-6023. [PMID: 32483434 PMCID: PMC7255011 DOI: 10.7150/thno.38742] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 04/22/2020] [Indexed: 12/22/2022] Open
Abstract
Therapeutic cancer vaccines are one of the most promising strategies of immunotherapy. Traditional vaccines consisting of tumor-associated antigens have met with limited success. Recently, neoantigens derived from nonsynonymous mutations in tumor cells have emerged as alternatives that can improve tumor-specificity and reduce on-target off-tumor toxicity. Synthetic peptides are a common platform for neoantigen vaccines. It has been suggested that extending short peptides into long peptides can overcome immune tolerance and induce both CD4+ and CD8+ T cell responses. This review will introduce the history of long peptide-based neoantigen vaccines, discuss their advantages, summarize current preclinical and clinical developments, and propose future perspectives.
Collapse
|
100
|
Lin W, Xu Y, Chen X, Liu J, Weng Y, Zhuang Q, Lin F, Huang Z, Wu S, Ding J, Chen L, Qiu X, Zhang L, Wu J, Lin D, Qiu S. Radiation-induced small extracellular vesicles as "carriages" promote tumor antigen release and trigger antitumor immunity. Am J Cancer Res 2020; 10:4871-4884. [PMID: 32308755 PMCID: PMC7163438 DOI: 10.7150/thno.43539] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/15/2020] [Indexed: 12/26/2022] Open
Abstract
Rationale: Accumulating evidence supports the importance of radiation therapy in the induction of antitumor immunity. Small extracellular vesicles (sEVs) play essential roles in tumor antigen loading and delivery. However, the role of sEVs in radiation-induced antitumor immunity remains unclear. It is therefore important to determine the role and regulatory mechanisms of sEVs in radiation-induced immunity. Methods: Tumor cells were irradiated (8 Gy), and sEVs were purified via ultracentrifugation. Primary tumor and experimental lung metastasis models were established in mice to evaluate antitumor immunity triggered by immunization with sEVs. Proteomic and bioinformatic analyses were performed to identify altered cargos in sEVs induced by radiation. Peptides derived from up-regulated proteins in sEVs were designed and synthesized as vaccines according to major histocompatibility complex (MHC) I binding and immunogenicity. Results: Here, we demonstrated that sEVs derived from irradiated tumor cells could trigger antitumor immunity against primary tumor and experimental lung metastasis by enhancing CD8+ and CD4+ T cell infiltration. Radiation may also enrich sEVs with tumor antigens and heat-shock proteins. Furthermore, CUB domain-containing protein 1 (CDCP1) derived from radiation-induced sEVs was identified as a novel tumor-associated antigen and developed as a peptide vaccine that may generate antitumor immune responses. Conclusions: Our results demonstrate that the use of sEVs secreted by irradiated tumor cells constitutes an efficient approach for tumor antigen delivery and presentation and highlight the role of sEVs in radiation-triggered antitumor immunity.
Collapse
|