1
|
Zou T, Huang Y, Zhou Z, He S, Liu J, Chen Y, Liu H, Luo Z, Liu M, Wei H, Yu C. A minimalist multifunctional nano-prodrug for drug resistance reverse and integration with PD-L1 mAb for enhanced immunotherapy of hepatocellular carcinoma. J Nanobiotechnology 2024; 22:750. [PMID: 39627819 PMCID: PMC11613529 DOI: 10.1186/s12951-024-03027-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/19/2024] [Indexed: 12/08/2024] Open
Abstract
Clinical treatment of hepatocellular carcinoma (HCC) with 5-fluorouracil (5-FU), the primary anticancer agent, remains unsatisfactory due to the glutathione (GSH)-associated drug resistance and immunosuppressive microenvironment of HCC. To develop a facile yet robust strategy to overcome 5-FU resistance for enhanced immunotherapy treatment of HCC via all dimensional GSH exhaustion, we report in this study construction of a minimalist prodrug consisting of 5-FU linked to an indoleamine-(2,3)-dioxygenase (IDO) inhibitor (IND) via a disulfide bridge, FU-SS-IND that can further self-assemble into stabilized nanoparticles, FU-SS-IND NPs. Specifically, besides the disulfide linker-induced GSH exhaustion, IND inhibits GSH biosynthesis and enhances the effector function of T cells for turning a "cold" tumor to a "hot" one, which synergistically achieving a tumor inhibition rate (TIR) of 92.5% in a 5-FU resistant mice model. Most importantly, FU-SS-IND NPs could upregulate programmed death ligand 1 (PD-L1) expression on the surface of tumor cells, which enables facile combination with immune checkpoint blockade (ICB) for a ultimate prolonged survival lifetime of 5-FU-resistant tumors-bearing mice. Overall, the minimalist bioreducible nano-prodrug developed herein demonstrates great translatable potential for efficiently reversing drug resistance and enhancing immunotherapy of HCC.
Collapse
Affiliation(s)
- Ting Zou
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yun Huang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Zongtao Zhou
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Shuangyan He
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jia Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yalan Chen
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Hongdu Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Zhonghui Luo
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Miaoxin Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science & MOE Key Lab of Rare Pediatric Disease, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - CuiYun Yu
- Affiliated Hospital of Hunan Academy of Chinese Medicine, Hunan Academy of Chinese Medicine, Changsha, 410013, China.
| |
Collapse
|
2
|
Thomas R, Jerome JM, Krieger KL, Ashraf N, Rowley DR. The reactive stroma response regulates the immune landscape in prostate cancer. JOURNAL OF TRANSLATIONAL GENETICS AND GENOMICS 2024; 8:249-77. [DOI: 10.20517/jtgg.2024.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Prostate cancer remains the most commonly diagnosed and the second leading cause of cancer-related deaths in men in the United States. The neoplastic transformation of prostate epithelia, concomitant with modulations in the stromal compartment, known as reactive stromal response, is critical for the growth, development, and progression of prostate cancer. Reactive stroma typifies an emergent response to disrupted tissue homeostasis commonly observed in wound repair and pathological conditions such as cancer. Despite the significance of reactive stroma in prostate cancer pathobiology, our understanding of the ontogeny, phenotypic and functional heterogeneity, and reactive stromal regulation of the immune microenvironment in prostate cancer remains limited. Traditionally characterized to have an immunologically "cold" tumor microenvironment, prostate cancer presents significant challenges for advancing immunotherapy compared to other solid tumors. This review explores the detrimental role of reactive stroma in prostate cancer, particularly its immunomodulatory function. Understanding the molecular characteristics and dynamic transcriptional program of the reactive stromal populations in tandem with tumor progression could offer insights into enhancing immunotherapy efficacy against prostate cancer.
Collapse
|
3
|
Chen C, Xu Y, Lai Z, Li Z, Sun Q. Case Report: Exploration of changes in serum immunoinflammation-related protein complexes of patients with metastatic breast cancer. Front Oncol 2023; 13:1207991. [PMID: 37546392 PMCID: PMC10401826 DOI: 10.3389/fonc.2023.1207991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/26/2023] [Indexed: 08/08/2023] Open
Abstract
Patients with advanced breast cancer are difficult to treat and have poor prognosis. At present, the commonly used methods to monitor the disease progression of breast cancer are imaging examinations such as breast ultrasound, mammography and peripheral blood tumor markers such as carcinoembryonic antigen (CEA) and carbohydrate antigen 15-3 (CA15-3). However, none of them can detect tumor progression at an early stage. Serum immunoinflammation-related protein complexes (IIRPCs) showed potential to indicate cancer progression. Therefore, we attempted to monitor the level of IIRPCs in peripheral blood of patients with metastatic breast cancer and compare it with patients' treatment and disease progression, and here we performed case reports of two of them.
Collapse
Affiliation(s)
- Chang Chen
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yali Xu
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhizhen Lai
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Zhili Li
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Qiang Sun
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Ligustilide Inhibits Tumor Angiogenesis by Downregulating VEGFA Secretion from Cancer-Associated Fibroblasts in Prostate Cancer via TLR4. Cancers (Basel) 2022; 14:cancers14102406. [PMID: 35626012 PMCID: PMC9140166 DOI: 10.3390/cancers14102406] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/30/2022] [Accepted: 05/10/2022] [Indexed: 12/05/2022] Open
Abstract
CAFs secrete VEGFA in the tumor microenvironment to induce angiogenesis and promote tumor growth. The downregulation of VEGFA secretion from CAFs helps block angiogenesis and exerts an anti-tumor effect. In vivo experiments showed that the angiogenesis of the tumor-bearing mice in the ligustilide group was significantly reduced. The results of MTT, tube formation, Transwell and scratch experiments showed that ligustilide did not affect the proliferation of HUVECs in a certain concentration range (<60 μM), but it inhibited the proliferation, tube formation and migration of HUVECs induced by CAFs. At this concentration, ligustilide did not inhibit CAF proliferation. The qPCR and WB results revealed that ligustilide downregulated the level of VEGFA in CAFs via the TLR4-ERK/JNK/p38 signaling pathway, and the effect was attenuated by blockers of the above molecules. Ligustilide also downregulated the autocrine VEGFA of HUVECs induced by CAFs, which inhibited angiogenesis more effectively. In addition, ligustilide inhibited glycolysis and HIF-1 expression in CAFs. Overall, ligustilide downregulated the VEGFA level in CAFs via the TLR4-ERK/JNK/p38 signaling pathway and inhibited the promotion of angiogenesis. This study provides a new strategy for the anti-tumor effect of natural active molecules, namely, blockade of angiogenesis, and provides a new candidate molecule for blocking angiogenesis in the tumor microenvironment.
Collapse
|
5
|
Jiang X, Wang X, Shen T, Yao Y, Chen M, Li Z, Li X, Shen J, Kou Y, Chen S, Zhou X, Luo Z, Cheng Z. FAPI-04 PET/CT Using [ 18F]AlF Labeling Strategy: Automatic Synthesis, Quality Control, and In Vivo Assessment in Patient. Front Oncol 2021; 11:649148. [PMID: 33816303 PMCID: PMC8017320 DOI: 10.3389/fonc.2021.649148] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 01/27/2021] [Indexed: 12/20/2022] Open
Abstract
68Ga labeled FAPI is the current standard for FAPI-PET, but its batch activity is limited. [18F]AlF-NOTA-FAPI-04 is a promising alternative combining the advantages of a chelator-based radiolabeling method with the unique properties of fluorine-18. The objective of this study was to develop a quick automatic method for synthesis of [18F]AlF-NOTA-FAPI-04 using a AllinOne synthesis system, and perform PET imaging with [18F]AlF-NOTA-FAPI-04 on patients. [18F]AlF-NOTA-FAPI-04 was produced, and its quality control was conducted by HPLC equipped with a radioactive detector. [18F]AlF-NOTA-FAPI-04 PET/CT imaging was performed in normal BALB/c mice (n = 3) and 4T1 breast cancer models (n = 3) to determine its biodistribution. Then [18F]AlF-NOTA-FAPI-04 and 18F-fluorodeoxyglucose (FDG) PET/CT imaging were performed in an invasive ductal carcinoma patient (female, 54 years old). The synthesis time of [18F]AlF-NOTA-FAPI-04 was about 25 min, and the radiochemical yield was 26.4 ± 1.5% (attenuation correction, n = 10). The radiochemical purity was above 99.0% and was above 98.0% after 6 h. The product was colorless transparent solution with pH value of 7.0-7.5, and the specific activity was 49.41 ± 3.19 GBq/μmol. PET/CT imaging in mice showed that physiological uptake of [18F]AlF-NOTA-FAPI-04 was mainly in the biliary system and bladder, and [18F]AlF-NOTA-FAPI-04 highly concentrated in tumor xenografts. PET/CT imaging in the patient showed that [18F]AlF-NOTA-FAPI-04 obtained high tumor background ratio (TBR) value of 8.44 in segment V and VI, while TBR value was 2.55 by 18F-FDG. [18F]AlF-NOTA-FAPI-04 could be synthesized with high radiochemical yield and batch production by AllinOne module and show excellent diagnosis performance in cancer patients.
Collapse
Affiliation(s)
- Xiao Jiang
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China.,Institute of Isotope, China Institute of Atomic Energy, Beijing, China
| | - Xiaoxiong Wang
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| | - Taipeng Shen
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| | - Yutang Yao
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| | - Meihua Chen
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| | - Zeng Li
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| | - Xiuli Li
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| | - Jiaqi Shen
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| | - Ying Kou
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| | - Shirong Chen
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| | - Xing Zhou
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| | - Zhifu Luo
- Institute of Isotope, China Institute of Atomic Energy, Beijing, China
| | - Zhuzhong Cheng
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| |
Collapse
|
6
|
Xie J, Yuan S, Peng L, Li H, Niu L, Xu H, Guo X, Yang M, Duan F. Antitumor immunity targeting fibroblast activation protein-α in a mouse Lewis lung carcinoma model. Oncol Lett 2020; 20:868-876. [PMID: 32566014 PMCID: PMC7285819 DOI: 10.3892/ol.2020.11637] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 04/15/2020] [Indexed: 12/12/2022] Open
Abstract
The tumor stromal microenvironment is an integral part of the occurrence and development of tumor. Cancer-associated fibroblasts (CAFs) are a key component of most tumor stromal microenvironments. The present study aimed to investigate the use of CAFs-targeted immunotherapy to fibroblast activation protein-α (FAP-α) expressed in CAFs. Recombinant adenoviral vectors containing the mouse FAP-α cDNA (rAd-FAP-α) were constructed. C57BL/6 mice were immunized with rAd-FAP-α infected dendritic cells (DCs) against FAP-α, which is overexpress in CAFs. The results demonstrated that mice vaccinated with rAd-FAP-α DCs gave rise to potent FAP-α-specific cytotoxic T lymphocytes capable of lysing Lewis lung cancer (LLC) CAFs. Furthermore, mice vaccinated with rAd-FAP-α-transduced DCs induced an effective therapeutic or protective antitumor immunity to LLC in a subcutaneous model, and prolonged overall survival time compared with mice vaccinated with the control recombinant adenovirus-transduced DCs (rAd-c DCs) or DCs alone. The results of the present study suggested that FAP-α, which is preferentially expressed in CAFs, may be considered as a potential target for killing or destroying CAFs within the tumor stromal microenvironment, and may be exploited to develop immunogenic tumor vaccines.
Collapse
Affiliation(s)
- Junping Xie
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shiyang Yuan
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Laishui Peng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Huanyu Li
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Linxia Niu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Hui Xu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiaolin Guo
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Mei Yang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Fengying Duan
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
7
|
MPSSS impairs the immunosuppressive function of cancer-associated fibroblasts via the TLR4-NF-κB pathway. Biosci Rep 2019; 39:BSR20182171. [PMID: 30992392 PMCID: PMC6509060 DOI: 10.1042/bsr20182171] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 03/29/2019] [Accepted: 04/09/2019] [Indexed: 02/06/2023] Open
Abstract
The polysaccharides MPSSS was extracted from Lentinus edodes and has been reported to effectively inhibit tumor growth and eliminate the function of myeloid-derived immune suppressor cell-mediated T cell inhibition, thus improving the efficacy of cancer therapy. The exploration of how MPSSS affects the functions of cancer-associated fibroblasts (CAFs) will provide a new perspective for understanding the antitumor effects of MPSSS. In the present study, prostate CAFs were selected as target cells to study whether MPSSS affected cell proliferation and function. The results showed that MPSSS did not directly inhibit the growth of prostate CAFs but interfered with CAF-mediated T cell inhibition and affected the immunosuppressive function of prostate CAFs. Mechanistic studies were further performed and showed that MPSSS activated key node proteins in the NF-κB pathway that were dependent on MyD88, and a TLR4 inhibitor blocked the changes in these proteins and the effect of MPSSS. We hypothesize that MPSSS can activate the MyD88-dependent TLR4-NF-κB signaling pathway to change the function of CAFs. In conclusion, these results demonstrate that MPSSS can not only effectively inhibit the growth of prostate cancer as we previously reported but also alter the function of prostate CAFs by activating the TLR4-NF-κB pathway, providing a new strategy for the comprehensive treatment of tumors.
Collapse
|
8
|
Ma J, Xu Y, Zheng Q, Wang Y, Hu M, Ma F, Long H, Qin Z, Tao N. Ligustilide inhibits the activation of cancer-associated fibroblasts. Life Sci 2019; 218:58-64. [DOI: 10.1016/j.lfs.2018.12.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 12/16/2018] [Accepted: 12/18/2018] [Indexed: 11/27/2022]
|
9
|
Zhang MX, Gan W, Jing CY, Zheng SS, Zhang J, Shen HJ, Xu X, Lin JJ, Zhang BH, Qiu SJ. Overexpression of interleukin-35 in intrahepatic cholangiocarcinoma is a prognostic indicator after curative resection. Cancer Sci 2018; 109:1195-1206. [PMID: 29446854 PMCID: PMC5891208 DOI: 10.1111/cas.13535] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/08/2018] [Accepted: 02/10/2018] [Indexed: 02/06/2023] Open
Abstract
Interleukin‐35 (IL‐35) is implicated in tumorigenesis, but its exact impact on intrahepatic cholangiocarcinoma (ICC) is not clear. The aim of the present study was to explore the specific effect of IL‐35 on patient prognosis. Additionally, we formulated an effective prognostic nomogram for ICC patients after curative resection. Immunohistochemistry was applied to explore IL‐35 expression as well as IL‐35 receptor (IL‐35R) in 102 ICC patients. Results showed that IL‐35 was highly expressed in ICC tumor tissues and was positively associated with lymph node metastasis (LNM), TNM stage and vascular invasion and was an independent prognostic factor for patients' overall survival (OS) and recurrence‐free survival (RFS). High expression of IL‐35R (gp130 and IL‐12Rβ2) was also observed in ICC cancer tissues, but only gp130 was an independent prognostic factor for OS and RFS and was indispensable in IL‐35‐mediated ICC clinical prognosis. The nomogram comprising carcinoembryonic antigen, LNM, IL‐35 and gp130 expression achieved better predictive accuracy compared with TNM stage for OS. Our data support that high IL‐35 expression correlates with ICC aggressiveness and emerges as a valuable biomarker for evaluating ICC progression and prognosis in clinical work.
Collapse
Affiliation(s)
- Mei-Xia Zhang
- Liver Cancer Institute and Zhongshan Hospital of Fudan University, Shanghai, China
| | - Wei Gan
- Liver Cancer Institute and Zhongshan Hospital of Fudan University, Shanghai, China
| | - Chu-Yu Jing
- Liver Cancer Institute and Zhongshan Hospital of Fudan University, Shanghai, China
| | - Su-Su Zheng
- Liver Cancer Institute and Zhongshan Hospital of Fudan University, Shanghai, China
| | - Juan Zhang
- Liver Cancer Institute and Zhongshan Hospital of Fudan University, Shanghai, China
| | - Hu-Jia Shen
- Liver Cancer Institute and Zhongshan Hospital of Fudan University, Shanghai, China
| | - Xin Xu
- Liver Cancer Institute and Zhongshan Hospital of Fudan University, Shanghai, China
| | - Jia-Jia Lin
- Liver Cancer Institute and Zhongshan Hospital of Fudan University, Shanghai, China
| | - Bo-Heng Zhang
- Liver Cancer Institute and Zhongshan Hospital of Fudan University, Shanghai, China.,Center for Evidence-Based Medicine, Fudan University, Shanghai, China
| | - Shuang-Jian Qiu
- Liver Cancer Institute and Zhongshan Hospital of Fudan University, Shanghai, China
| |
Collapse
|
10
|
Chen M, Lei X, Shi C, Huang M, Li X, Wu B, Li Z, Han W, Du B, Hu J, Nie Q, Mai W, Ma N, Xu N, Zhang X, Fan C, Hong A, Xia M, Luo L, Ma A, Li H, Yu Q, Chen H, Zhang D, Ye W. Pericyte-targeting prodrug overcomes tumor resistance to vascular disrupting agents. J Clin Invest 2017; 127:3689-3701. [PMID: 28846068 DOI: 10.1172/jci94258] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/11/2017] [Indexed: 01/10/2023] Open
Abstract
Blood vessels in the tumor periphery have high pericyte coverage and are resistant to vascular disrupting agents (VDAs). VDA treatment resistance leads to a viable peripheral tumor rim that contributes to treatment failure and disease recurrence. Here, we provide evidence to support a hypothesis that shifting the target of VDAs from tumor vessel endothelial cells to pericytes disrupts tumor peripheral vessels and the viable rim, circumventing VDA treatment resistance. Through chemical engineering, we developed Z-GP-DAVLBH (from the tubulin-binding VDA desacetylvinblastine monohydrazide [DAVLBH]) as a prodrug that can be selectively activated by fibroblast activation protein α (FAPα) in tumor pericytes. Z-GP-DAVLBH selectively destroys the cytoskeleton of FAPα-expressing tumor pericytes, disrupting blood vessels both within the core and around the periphery of tumors. As a result, Z-GP-DAVLBH treatment eradicated the otherwise VDA-resistant tumor rim and led to complete regression of tumors in multiple lines of xenografts without producing the drug-related toxicity that is associated with similar doses of DAVLBH. This study demonstrates that targeting tumor pericytes with an FAPα-activated VDA prodrug represents a potential vascular disruption strategy in overcoming tumor resistance to VDA treatments.
Collapse
Affiliation(s)
- Minfeng Chen
- College of Pharmacy, and.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Xueping Lei
- College of Pharmacy, and.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Changzheng Shi
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Maohua Huang
- College of Pharmacy, and.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Xiaobo Li
- College of Pharmacy, and.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Baojian Wu
- College of Pharmacy, and.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Zhengqiu Li
- College of Pharmacy, and.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Weili Han
- School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, China
| | - Bin Du
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jianyang Hu
- College of Pharmacy, and.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Qiulin Nie
- College of Pharmacy, and.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Weiqian Mai
- College of Pharmacy, and.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Nan Ma
- College of Pharmacy, and.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Nanhui Xu
- College of Pharmacy, and.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Xinyi Zhang
- College of Pharmacy, and.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Chunlin Fan
- College of Pharmacy, and.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Aihua Hong
- Analytical and Testing Center, Jinan University, Guangzhou, China
| | - Minghan Xia
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Liangping Luo
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ande Ma
- School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, China
| | - Hongsheng Li
- Cancer Center of Guangzhou Medical University, Guangzhou, China
| | - Qiang Yu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Heru Chen
- College of Pharmacy, and.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Dongmei Zhang
- College of Pharmacy, and.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| | - Wencai Ye
- College of Pharmacy, and.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, China
| |
Collapse
|
11
|
Tan SY, Chowdhury S, Polak N, Gorrell MD, Weninger W. Fibroblast activation protein is dispensable in the anti-influenza immune response in mice. PLoS One 2017; 12:e0171194. [PMID: 28158223 PMCID: PMC5291439 DOI: 10.1371/journal.pone.0171194] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 01/18/2017] [Indexed: 01/10/2023] Open
Abstract
Fibroblast activation protein alpha (FAP) is a unique dual peptidase of the S9B serine protease family, being capable of both dipeptidyl peptidase and endopeptidase activities. FAP is expressed at low level in healthy adult organs including the pancreas, cervix, uterus, submaxillary gland and the skin, and highly upregulated in embryogenesis, chronic inflammation and tissue remodelling. It is also expressed by cancer-associated stromal fibroblasts in more than 90% of epithelial tumours. FAP has enzymatic and non-enzymatic functions in the growth, immunosuppression, invasion and cell signalling of tumour cells. FAP deficient mice are fertile and viable with no gross abnormality, but little data exist on the role of FAP in the immune system. FAP is upregulated in association with microbial stimulation and chronic inflammation, but its function in infection remains unknown. We showed that major populations of immune cells including CD4+ and CD8+ T cells, B cells, dendritic cells and neutrophils are generated and maintained normally in FAP knockout mice. Upon intranasal challenge with influenza virus, FAP mRNA was increased in the lungs and lung-draining lymph nodes. Nonetheless, FAP deficient mice showed similar pathologic kinetics to wildtype controls, and were capable of supporting normal anti-influenza T and B cell responses. There was no evidence of compensatory upregulation of other DPP4 family members in influenza-infected FAP-deficient mice. FAP appears to be dispensable in anti-influenza adaptive immunity.
Collapse
Affiliation(s)
- Sioh-Yang Tan
- Immune Imaging Program, Centenary Institute for Cancer Medicine and Cell Biology, Newtown, New South Wales, Australia
- Sydney Medical School, The University of Sydney, New South Wales, Australia
| | - Sumaiya Chowdhury
- Sydney Medical School, The University of Sydney, New South Wales, Australia
- Molecular Hepatology Laboratory, Centenary Institute for Cancer Medicine and Cell Biology, Newtown, New South Wales, Australia
| | - Natasa Polak
- Sydney Medical School, The University of Sydney, New South Wales, Australia
- Molecular Hepatology Laboratory, Centenary Institute for Cancer Medicine and Cell Biology, Newtown, New South Wales, Australia
| | - Mark D. Gorrell
- Sydney Medical School, The University of Sydney, New South Wales, Australia
- Molecular Hepatology Laboratory, Centenary Institute for Cancer Medicine and Cell Biology, Newtown, New South Wales, Australia
| | - Wolfgang Weninger
- Immune Imaging Program, Centenary Institute for Cancer Medicine and Cell Biology, Newtown, New South Wales, Australia
- Sydney Medical School, The University of Sydney, New South Wales, Australia
- Department of Dermatology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| |
Collapse
|
12
|
A whole-cell tumor vaccine modified to express fibroblast activation protein induces antitumor immunity against both tumor cells and cancer-associated fibroblasts. Sci Rep 2015; 5:14421. [PMID: 26394925 PMCID: PMC4585784 DOI: 10.1038/srep14421] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 08/19/2015] [Indexed: 02/05/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) are common components of the tumor-suppressive microenvironment, and are a major determinant of the poor outcome of therapeutic vaccination. In this study, we modified tumor cells to express the fibroblast activation protein (FAP), which is highly expressed by CAFs, to potentially improve whole-cell tumor vaccines by targeting both tumor cells and CAFs. Tumor cells were transfected with murine FAP plasmids bearing the cationic lipid DOTAP. Its antitumor effects were investigated in three established tumor models. Vaccination with tumor cells expressing FAP eliminated solid tumors and tumors resulting from hematogenous dissemination. This antitumor immune response was mediated by CD8+ T cells. Additionally, we found that CAFs were significantly reduced within the tumors. Furthermore, this vaccine enhanced the infiltration of CD8+ T lymphocytes, and suppressed the accumulation of immunosuppressive cells in the tumor microenvironment. Our results indicated that the FAP-modified whole-cell tumor vaccine induced strong antitumor immunity against both tumor cells and CAFs and reversed the immunosuppressive effects of tumors by decreasing the recruitment of immunosuppressive cells and enhancing the recruitment of effector T cells. This conclusion may have important implications for the clinical use of genetically modified tumor cells as cancer vaccines.
Collapse
|
13
|
Liao R, Tang ZW, Li DW, Luo SQ, Huang P, Du CY. Preoperative neutrophil-to-lymphocyte ratio predicts recurrence of patients with single-nodule small hepatocellular carcinoma following curative resection: a retrospective report. World J Surg Oncol 2015; 13:265. [PMID: 26328917 PMCID: PMC4557750 DOI: 10.1186/s12957-015-0670-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 08/03/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Preoperative neutrophil-to-lymphocyte ratio (NLR) has been identified as a predictor for the recurrence of hepatocellular carcinoma (HCC), but the cut-off of NLR is inconsistent in various studies. Thus, we detected the prognostic value of preoperative NLR in the single-nodule small HCC (SHCC) patients using X-tile for cutpoint. METHODS Between January 2007 and December 2010, a total of 222 single-nodule SHCC patients underwent curative resection and were examined for the prognostic roles of preoperative NLR by X-tile. RESULTS In this study, all patients were divided into the low-NLR subgroup (NLR ≤ 2.1) and the high-NLR subgroup (NLR > 2.1) by X-tile. Preoperative NLR showed predictive value for time to recurrence (TTR) and overall survival (OS). Moreover, NLR was associated with total bilirubin, white blood cell counts, and HBsAg, respectively (P = 0.012, <0.001, and 0.011, respectively). Especially, NLR could discriminate the outcome of patients in the subgroup with alpha-fetoprotein (AFP) levels of ≤400 ng/mL. Importantly, postoperative transcatheter arterial chemoembolization (TACE) had close relationship with OS (P = 0.001) and TTR (P ≤ 0.001). CONCLUSIONS Therefore, this study indicates that preoperative NLR, divided by X-tile for the cutpoint, is a simple prognostic marker for the patients with single-nodule SHCC after curative resection.
Collapse
Affiliation(s)
- Rui Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Zhuo-Wei Tang
- Department of General Surgery, Mianyang Central Hospital, Mianyang, 621000, China.
| | - De-Wei Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Shi-Qiao Luo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Ping Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Cheng-You Du
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
14
|
Smith AD, Roda D, Yap TA. Strategies for modern biomarker and drug development in oncology. J Hematol Oncol 2014; 7:70. [PMID: 25277503 PMCID: PMC4189730 DOI: 10.1186/s13045-014-0070-8] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 09/21/2014] [Indexed: 02/08/2023] Open
Abstract
Technological advancements in the molecular characterization of cancers have enabled researchers to identify an increasing number of key molecular drivers of cancer progression. These discoveries have led to multiple novel anticancer therapeutics, and clinical benefit in selected patient populations. Despite this, the identification of clinically relevant predictive biomarkers of response continues to lag behind. In this review, we discuss strategies for the molecular characterization of cancers and the importance of biomarkers for the development of novel antitumor therapeutics. We also review critical successes and failures in oncology, and detail the lessons learnt, which may aid in the acceleration of anticancer drug development and biomarker discovery.
Collapse
Affiliation(s)
- Alan D Smith
- Drug Development Unit, Royal Marsden NHS Foundation Trust, Division of Clinical Studies, The Institute of Cancer Research, Downs Road, Sutton, Surrey, SM2 5PT, UK.
| | - Desam Roda
- Drug Development Unit, Royal Marsden NHS Foundation Trust, Division of Clinical Studies, The Institute of Cancer Research, Downs Road, Sutton, Surrey, SM2 5PT, UK.
| | - Timothy A Yap
- Drug Development Unit, Royal Marsden NHS Foundation Trust, Division of Clinical Studies, The Institute of Cancer Research, Downs Road, Sutton, Surrey, SM2 5PT, UK.
| |
Collapse
|
15
|
Wang Y, Song G, Wang Y, Qiu L, Qin X, Liu H, Li F, Wang X, Li F, Guo S, Zhang Y, Li Z. Elevated Serum Levels of Circulating Immunoinflammation-Related Protein Complexes Are Associated with Cancer. J Proteome Res 2013; 13:710-9. [DOI: 10.1021/pr4008255] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Yanying Wang
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing 100005, P. R. China
| | - Gaoguang Song
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing 100005, P. R. China
| | - Yanmin Wang
- Department
of Clinical Laboratory, Heze Municipal Hospital, Shandong 274031, P. R. China
| | - Ling Qiu
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, P. R. China
| | - Xuzhen Qin
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, P. R. China
| | - Hui Liu
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing 100005, P. R. China
| | - Fang Li
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing 100005, P. R. China
| | - Xiaodong Wang
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing 100005, P. R. China
| | - Fenjie Li
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing 100005, P. R. China
| | - Shuai Guo
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing 100005, P. R. China
| | - Yaping Zhang
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing 100005, P. R. China
| | - Zhili Li
- Department of Biophysics and Structural Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing 100005, P. R. China
| |
Collapse
|
16
|
Wu Y, Garmire LX, Fan R. Inter-cellular signaling network reveals a mechanistic transition in tumor microenvironment. Integr Biol (Camb) 2013; 4:1478-86. [PMID: 23080410 DOI: 10.1039/c2ib20044a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
We conducted inter-cellular cytokine correlation and network analysis based upon a stochastic population dynamics model that comprises five cell types and fifteen signaling molecules inter-connected through a large number of cell-cell communication pathways. We observed that the signaling molecules are tightly correlated even at very early stages (e.g. the first month) of human glioma, but such correlation rapidly diminishes when tumor grows to a size that can be clinically detected. Further analysis suggests that paracrine is shown to be the dominant force during tumor initiation and priming, while autocrine supersedes it and supports a robust tumor expansion. In correspondence, the cytokine correlation network evolves through an increasing to decreasing complexity. This study indicates a possible mechanistic transition from the microenvironment-controlled, paracrine-based regulatory mechanism to self-sustained rapid progression to fetal malignancy. It also reveals key nodes that are responsible for such transition and can be potentially harnessed for the design of new anti-cancer therapies.
Collapse
Affiliation(s)
- Yu Wu
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | | | | |
Collapse
|
17
|
Scarlett CJ. Contribution of bone marrow derived cells to the pancreatic tumor microenvironment. Front Physiol 2013; 4:56. [PMID: 23531764 PMCID: PMC3607802 DOI: 10.3389/fphys.2013.00056] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 03/08/2013] [Indexed: 12/18/2022] Open
Abstract
Pancreatic cancer is a complex, aggressive, and heterogeneous malignancy driven by the multifaceted interactions within the tumor microenvironment. While it is known that the tumor microenvironment accommodates many cell types, each playing a key role in tumorigenesis, the major source of these stromal cells is not well-understood. This review examines the contribution of bone marrow-derived cells (BMDC) to pancreatic carcinogenesis, with respect to their role in constituting the tumor microenvironment. In particular, their role in supporting fibrosis, immunosuppression, and neovascularization will be discussed.
Collapse
Affiliation(s)
- Christopher J Scarlett
- Food Bioactives and Pancreatic Cancer Biology Group, School of Environmental and Life Sciences, University of Newcastle Ourimbah, NSW, Australia ; Cancer Research Program, Garvan Institute of Medical Research Darlinghurst, Sydney, NSW, Australia
| |
Collapse
|
18
|
Johnson TS, Munn DH. Host Indoleamine 2,3-Dioxygenase: Contribution to Systemic Acquired Tumor Tolerance. Immunol Invest 2012; 41:765-97. [DOI: 10.3109/08820139.2012.689405] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
19
|
In silico experimentation of glioma microenvironment development and anti-tumor therapy. PLoS Comput Biol 2012; 8:e1002355. [PMID: 22319429 PMCID: PMC3271023 DOI: 10.1371/journal.pcbi.1002355] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 12/04/2011] [Indexed: 12/31/2022] Open
Abstract
Tumor cells do not develop in isolation, but co-evolve with stromal cells and tumor-associated immune cells in a tumor microenvironment mediated by an array of soluble factors, forming a complex intercellular signaling network. Herein, we report an unbiased, generic model to integrate prior biochemical data and the constructed brain tumor microenvironment in silico as characterized by an intercellular signaling network comprising 5 types of cells, 15 cytokines, and 69 signaling pathways. The results show that glioma develops through three distinct phases: pre-tumor, rapid expansion, and saturation. We designed a microglia depletion therapy and observed significant benefit for virtual patients treated at the early stages but strikingly no therapeutic efficacy at all when therapy was given at a slightly later stage. Cytokine combination therapy exhibits more focused and enhanced therapeutic response even when microglia depletion therapy already fails. It was further revealed that the optimal combination depends on the molecular profile of individual patients, suggesting the need for patient stratification and personalized treatment. These results, obtained solely by observing the in silico dynamics of the glioma microenvironment with no fitting to experimental/clinical data, reflect many characteristics of human glioma development and imply new venues for treating tumors via selective targeting of microenvironmental components.
Collapse
|
20
|
Modulation of tumor tolerance in primary central nervous system malignancies. Clin Dev Immunol 2012; 2012:937253. [PMID: 22312408 PMCID: PMC3270544 DOI: 10.1155/2012/937253] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2011] [Revised: 09/29/2011] [Accepted: 10/03/2011] [Indexed: 12/14/2022]
Abstract
Central nervous system tumors take advantage of the unique immunology of the CNS and develop exquisitely complex stromal networks that promote growth despite the presence of antigen-presenting cells and tumor-infiltrating lymphocytes. It is precisely this immunological paradox that is essential to the survival of the tumor. We review the evidence for functional CNS immune privilege and the impact it has on tumor tolerance. In this paper, we place an emphasis on the role of tumor-infiltrating myeloid cells in maintaining stromal and vascular quiescence, and we underscore the importance of indoleamine 2,3-dioxygenase activity as a myeloid-driven tumor tolerance mechanism. Much remains to be discovered regarding the tolerogenic mechanisms by which CNS tumors avoid immune clearance. Thus, it is an open question whether tumor tolerance in the brain is fundamentally different from that of peripheral sites of tumorigenesis or whether it simply stands as a particularly strong example of such tolerance.
Collapse
|
21
|
Scarlett CJ, Colvin EK, Pinese M, Chang DK, Morey AL, Musgrove EA, Pajic M, Apte M, Henshall SM, Sutherland RL, Kench JG, Biankin AV. Recruitment and activation of pancreatic stellate cells from the bone marrow in pancreatic cancer: a model of tumor-host interaction. PLoS One 2011; 6:e26088. [PMID: 22022519 PMCID: PMC3193536 DOI: 10.1371/journal.pone.0026088] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Accepted: 09/19/2011] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND AND AIMS Chronic pancreatitis and pancreatic cancer are characterised by extensive stellate cell mediated fibrosis, and current therapeutic development includes targeting pancreatic cancer stroma and tumor-host interactions. Recent evidence has suggested that circulating bone marrow derived stem cells (BMDC) contribute to solid organs. We aimed to define the role of circulating haematopoietic cells in the normal and diseased pancreas. METHODS Whole bone marrow was harvested from male β-actin-EGFP donor mice and transplanted into irradiated female recipient C57/BL6 mice. Chronic pancreatitis was induced with repeat injections of caerulein, while carcinogenesis was induced with an intrapancreatic injection of dimethylbenzanthracene (DMBA). Phenotype of engrafted donor-derived cells within the pancreas was assessed by immunohistochemistry, immunofluorescence and in situ hybridisation. RESULTS GFP positive cells were visible in the exocrine pancreatic epithelia from 3 months post transplantation. These exhibited acinar morphology and were positive for amylase and peanut agglutinin. Mice administered caerulein developed chronic pancreatitis while DMBA mice exhibited precursor lesions and pancreatic cancer. No acinar cells were identified to be donor-derived upon cessation of cerulein treatment, however rare occurrences of bone marrow-derived acinar cells were observed during pancreatic regeneration. Increased recruitment of BMDC was observed within the desmoplastic stroma, contributing to the activated pancreatic stellate cell (PaSC) population in both diseases. Expression of stellate cell markers CELSR3, PBX1 and GFAP was observed in BMD cancer-associated PaSCs, however cancer-associated, but not pancreatitis-associated BMD PaSCs, expressed the cancer PaSC specific marker CELSR3. CONCLUSIONS This study demonstrates that BMDC can incorporate into the pancreas and adopt the differentiated state of the exocrine compartment. BMDC that contribute to the activated PaSC population in chronic pancreatitis and pancreatic cancer have different phenotypes, and may play important roles in these diseases. Further, bone marrow transplantation may provide a useful model for the study of tumor-host interactions in cancer and pancreatitis.
Collapse
Affiliation(s)
- Christopher J. Scarlett
- Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia
| | - Emily K. Colvin
- Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia
| | - Mark Pinese
- Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia
| | - David K. Chang
- Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia
- Division of Surgery, Bankstown Hospital, Eldridge Road, Bankstown, Sydney, Australia
| | - Adrienne L. Morey
- Department of Anatomical Pathology, St Vincent's Hospital, Darlinghurst, Australia
| | - Elizabeth A. Musgrove
- Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia
| | - Marina Pajic
- Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia
| | - Minoti Apte
- South Western Sydney Clinical School, The University of New South Wales, Sydney, Australia
| | - Susan M. Henshall
- Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia
| | - Robert L. Sutherland
- Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia
| | - James G. Kench
- Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia
- Department of Anatomical Pathology, Royal Prince Alfred Hospital, Camperdown, Sydney, Australia
| | - Andrew V. Biankin
- Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia
- Division of Surgery, Bankstown Hospital, Eldridge Road, Bankstown, Sydney, Australia
| |
Collapse
|
22
|
Stephan MT, Irvine DJ. Enhancing Cell therapies from the Outside In: Cell Surface Engineering Using Synthetic Nanomaterials. NANO TODAY 2011; 6:309-325. [PMID: 21826117 PMCID: PMC3148657 DOI: 10.1016/j.nantod.2011.04.001] [Citation(s) in RCA: 178] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Therapeutic treatments based on the injection of living cells are in clinical use and preclinical development for diseases ranging from cancer to cardiovascular disease to diabetes. To enhance the function of therapeutic cells, a variety of chemical and materials science strategies are being developed that engineer the surface of therapeutic cells with new molecules, artificial receptors, and multifunctional nanomaterials, synthetically endowing donor cells with new properties and functions. These approaches offer a powerful complement to traditional genetic engineering strategies for enhancing the function of living cells.
Collapse
Affiliation(s)
- Matthias T. Stephan
- Department of Material Science and Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts, USA
| | - Darrell J. Irvine
- Department of Material Science and Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Ragon Institute of Massachusetts General Hospital, MIT and Harvard University, Boston, Massachusetts, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| |
Collapse
|