1
|
Shi J, Han W, Wang J, Kong X. Anti-Tumor Strategies Targeting Nutritional Deprivation: Challenges and Opportunities. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2415550. [PMID: 39895165 DOI: 10.1002/adma.202415550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 01/04/2025] [Indexed: 02/04/2025]
Abstract
Higher and richer nutrient requirements are typical features that distinguish tumor cells from AU: cells, ensuring adequate substrates and energy sources for tumor cell proliferation and migration. Therefore, nutrient deprivation strategies based on targeted technologies can induce impaired cell viability in tumor cells, which are more sensitive than normal cells. In this review, nutrients that are required by tumor cells and related metabolic pathways are introduced, and anti-tumor strategies developed to target nutrient deprivation are described. In addition to tumor cells, the nutritional and metabolic characteristics of other cells in the tumor microenvironment (including macrophages, neutrophils, natural killer cells, T cells, and cancer-associated fibroblasts) and related new anti-tumor strategies are also summarized. In conclusion, recent advances in anti-tumor strategies targeting nutrient blockade are reviewed, and the challenges and prospects of these anti-tumor strategies are discussed, which are of theoretical significance for optimizing the clinical application of tumor nutrition deprivation strategies.
Collapse
Affiliation(s)
- Jinsheng Shi
- Qingdao Key Lab of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266000, China
| | - Wei Han
- Qingdao Key Lab of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266000, China
| | - Jie Wang
- Pharmacy Department, Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital), Qingdao, Shandong, 266000, China
| | - Xiaoying Kong
- Institute of Regenerative Medicine and Laboratory Technology Innovation, Qingdao University, Qingdao, Shandong, 266071, China
| |
Collapse
|
2
|
Hu X, Ye K, Bo S, Xiao Z, Ma M, Pan J, Zhong X, Zhang D, Mo X, Yu X, Chen M, Luo L, Shi C. Monitoring imatinib decreasing pericyte coverage and HIF-1α level in a colorectal cancer model by an ultrahigh-field multiparametric MRI approach. J Transl Med 2024; 22:712. [PMID: 39085929 PMCID: PMC11293104 DOI: 10.1186/s12967-024-05497-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Excessive pericyte coverage promotes tumor growth, and a downregulation may solve this dilemma. Due to the double-edged sword role of vascular pericytes in tumor microenvironment (TME), indiscriminately decreasing pericyte coverage by imatinib causes poor treatment outcomes. Here, we optimized the use of imatinib in a colorectal cancer (CRC) model in high pericyte-coverage status, and revealed the value of multiparametric magnetic resonance imaging (mpMRI) at 9.4T in monitoring treatment-related changes in pericyte coverage and the TME. METHODS CRC xenograft models were evaluated by histological vascular characterizations and mpMRI. Mice with the highest pericyte coverage were treated with imatinib or saline; then, vascular characterizations, tumor apoptosis and HIF-1α level were analyzed histologically, and alterations in the expression of Bcl-2/bax pathway were assessed through qPCR. The effects of imatinib were monitored by dynamic contrast-enhanced (DCE)-, diffusion-weighted imaging (DWI)- and amide proton transfer chemical exchange saturation transfer (APT CEST)-MRI at 9.4T. RESULTS The DCE- parameters provided a good histologic match the tumor vascular characterizations. In the high pericyte coverage status, imatinib exhibited significant tumor growth inhibition, necrosis increase and pericyte coverage downregulation, and these changes were accompanied by increased vessel permeability, decreased microvessel density (MVD), increased tumor apoptosis and altered gene expression of apoptosis-related Bcl-2/bax pathway. Strategically, a 4-day imatinib effectively decreased pericyte coverage and HIF-1α level, and continuous treatment led to a less marked decrease in pericyte coverage and re-elevated HIF-1α level. Correlation analysis confirmed the feasibility of using mpMRI parameters to monitor imatinib treatment, with DCE-derived Ve and Ktrans being most correlated with pericyte coverage, Ve with vessel permeability, AUC with microvessel density (MVD), DWI-derived ADC with tumor apoptosis, and APT CEST-derived MTRasym at 1 µT with HIF-1α. CONCLUSIONS These results provided an optimized imatinib regimen to achieve decreasing pericyte coverage and HIF-1α level in the high pericyte-coverage CRC model, and offered an ultrahigh-field multiparametric MRI approach for monitoring pericyte coverage and dynamics response of the TME to treatment.
Collapse
Affiliation(s)
- Xinpeng Hu
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Kunlin Ye
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Shaowei Bo
- Department of Medical Imaging, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Xingang Middle Road No. 466, Guangzhou, 510317, China
| | - Zeyu Xiao
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
- Engineering Research Center of Medical Imaging Artificial Intelligence for Precision Diagnosis and Treatment, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Mengjie Ma
- Department of Radiology, Guangzhou First People's Hospital, Panfu Road No. 1, Guangzhou, 510080, China
| | - Jinghua Pan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Xing Zhong
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Dong Zhang
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Xukai Mo
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Xiaojun Yu
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China
| | - Minfeng Chen
- College of Pharmacy, Jinan University, West Huangpu Avenue No.601, Guangzhou, 510632, China.
| | - Liangping Luo
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China.
- Engineering Research Center of Medical Imaging Artificial Intelligence for Precision Diagnosis and Treatment, West Huangpu Avenue No. 613, Guangzhou, 510630, China.
| | - Changzheng Shi
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, West Huangpu Avenue No. 613, Guangzhou, 510630, China.
- Engineering Research Center of Medical Imaging Artificial Intelligence for Precision Diagnosis and Treatment, West Huangpu Avenue No. 613, Guangzhou, 510630, China.
| |
Collapse
|
3
|
Fan S, Qi M, Qi Q, Miao Q, Deng L, Pan J, Qiu S, He J, Huang M, Li X, Huang J, Lin J, Lyu W, Deng W, He Y, Liu X, Gao L, Zhang D, Ye W, Chen M. Targeting FAP α-positive lymph node metastatic tumor cells suppresses colorectal cancer metastasis. Acta Pharm Sin B 2024; 14:682-697. [PMID: 38322324 PMCID: PMC10840431 DOI: 10.1016/j.apsb.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/18/2023] [Accepted: 10/24/2023] [Indexed: 02/08/2024] Open
Abstract
Lymphatic metastasis is the main metastatic route for colorectal cancer, which increases the risk of cancer recurrence and distant metastasis. The properties of the lymph node metastatic colorectal cancer (LNM-CRC) cells are poorly understood, and effective therapies are still lacking. Here, we found that hypoxia-induced fibroblast activation protein alpha (FAPα) expression in LNM-CRC cells. Gain- or loss-function experiments demonstrated that FAPα enhanced tumor cell migration, invasion, epithelial-mesenchymal transition, stemness, and lymphangiogenesis via activation of the STAT3 pathway. In addition, FAPα in tumor cells induced extracellular matrix remodeling and established an immunosuppressive environment via recruiting regulatory T cells, to promote colorectal cancer lymph node metastasis (CRCLNM). Z-GP-DAVLBH, a FAPα-activated prodrug, inhibited CRCLNM by targeting FAPα-positive LNM-CRC cells. Our study highlights the role of FAPα in tumor cells in CRCLNM and provides a potential therapeutic target and promising strategy for CRCLNM.
Collapse
Affiliation(s)
- Shuran Fan
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Ming Qi
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Qi Qi
- School of Medicine, Jinan University, Guangzhou 510632, China
| | - Qun Miao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Lijuan Deng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Jinghua Pan
- The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Shenghui Qiu
- The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Jiashuai He
- The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Maohua Huang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Xiaobo Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Jie Huang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Jiapeng Lin
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Wenyu Lyu
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Weiqing Deng
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Yingyin He
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Xuesong Liu
- The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Lvfen Gao
- The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Dongmei Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Wencai Ye
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Minfeng Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Shenzhen 518055, China
| |
Collapse
|
4
|
Pellow C, Jafari Sojahrood A, Zhao X, Kolios MC, Exner AA, Goertz DE. Synchronous Intravital Imaging and Cavitation Monitoring of Antivascular Focused Ultrasound in Tumor Microvasculature Using Monodisperse Low Boiling Point Nanodroplets. ACS NANO 2024; 18:410-427. [PMID: 38147452 PMCID: PMC10786165 DOI: 10.1021/acsnano.3c07711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 12/28/2023]
Abstract
Focused ultrasound-stimulated microbubbles can induce blood flow shutdown and ischemic necrosis at higher pressures in an approach termed antivascular ultrasound. Combined with conventional therapies of chemotherapy, immunotherapy, and radiation therapy, this approach has demonstrated tumor growth inhibition and profound synergistic antitumor effects. However, the lower cavitation threshold of microbubbles can potentially yield off-target damage that the polydispersity of clinical agent may further exacerbate. Here we investigate the use of a monodisperse nanodroplet formulation for achieving antivascular effects in tumors. We first develop stable low boiling point monodisperse lipid nanodroplets and examine them as an alternative agent to mediate antivascular ultrasound. With synchronous intravital imaging and ultrasound monitoring of focused ultrasound-stimulated nanodroplets in tumor microvasculature, we show that nanodroplets can trigger blood flow shutdown and do so with a sharper pressure threshold and with fewer additional events than conventionally used microbubbles. We further leverage the smaller size and prolonged pharmacokinetic profile of nanodroplets to allow for potential passive accumulation in tumor tissue prior to antivascular ultrasound, which may be a means by which to promote selective tumor targeting. We find that vascular shutdown is accompanied by inertial cavitation and complex-order sub- and ultraharmonic acoustic signatures, presenting an opportunity for effective feedback control of antivascular ultrasound.
Collapse
Affiliation(s)
- Carly Pellow
- Sunnybrook
Research Institute, Toronto M4N 3M5, Canada
| | - Amin Jafari Sojahrood
- Sunnybrook
Research Institute, Toronto M4N 3M5, Canada
- Department
of Physics, Toronto Metropolitan University, Toronto M5B 2K3, Canada
- Institute
for Biomedical Engineering, Science and Technology (iBEST), a partnership
between St. Michael’s Hospital, a site of Unity Health Toronto
and Toronto Metropolitan University, Toronto M5B 1T8, Canada
| | - Xiaoxiao Zhao
- Sunnybrook
Research Institute, Toronto M4N 3M5, Canada
- Department
of Medical Biophysics, University of Toronto, Toronto M5G 1L7, Canada
| | - Michael C. Kolios
- Department
of Physics, Toronto Metropolitan University, Toronto M5B 2K3, Canada
- Institute
for Biomedical Engineering, Science and Technology (iBEST), a partnership
between St. Michael’s Hospital, a site of Unity Health Toronto
and Toronto Metropolitan University, Toronto M5B 1T8, Canada
| | - Agata A. Exner
- Department
of Radiology, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - David E. Goertz
- Sunnybrook
Research Institute, Toronto M4N 3M5, Canada
- Department
of Medical Biophysics, University of Toronto, Toronto M5G 1L7, Canada
| |
Collapse
|
5
|
Wang C, Huang M, Lin Y, Zhang Y, Pan J, Jiang C, Cheng M, Li S, He W, Li Z, Tu Z, Fan J, Zeng H, Lin J, Wang Y, Yao N, Liu T, Qi Q, Liu X, Zhang Z, Chen M, Xia L, Zhang D, Ye W. ENO2-derived phosphoenolpyruvate functions as an endogenous inhibitor of HDAC1 and confers resistance to antiangiogenic therapy. Nat Metab 2023; 5:1765-1786. [PMID: 37667133 DOI: 10.1038/s42255-023-00883-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/31/2023] [Indexed: 09/06/2023]
Abstract
Metabolic reprogramming is associated with resistance to antiangiogenic therapy in cancer. However, its molecular mechanisms have not been clearly elucidated. Here, we identify the glycolytic enzyme enolase 2 (ENO2) as a driver of resistance to antiangiogenic therapy in colorectal cancer (CRC) mouse models and human participants. ENO2 overexpression induces neuroendocrine differentiation, promotes malignant behaviour in CRC and desensitizes CRC to antiangiogenic drugs. Mechanistically, the ENO2-derived metabolite phosphoenolpyruvate (PEP) selectively inhibits histone deacetylase 1 (HDAC1) activity, which increases the acetylation of β-catenin and activates the β-catenin pathway in CRC. Inhibition of ENO2 with enolase inhibitors AP-III-a4 or POMHEX synergizes the efficacy of antiangiogenic drugs in vitro and in mice bearing drug-resistant CRC xenograft tumours. Together, our findings reveal that ENO2 constitutes a useful predictive biomarker and therapeutic target for resistance to antiangiogenic therapy in CRC, and uncover a previously undefined and metabolism-independent role of PEP in regulating resistance to antiangiogenic therapy by functioning as an endogenous HDAC1 inhibitor.
Collapse
Affiliation(s)
- Chenran Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- The First Affiliated Hospital of Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Maohua Huang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Yuning Lin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Yiming Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Jinghua Pan
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Chang Jiang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Minjing Cheng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Shenrong Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Wenzhuo He
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zhengqiu Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Zhengchao Tu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Jun Fan
- School of Medicine, Jinan University, Guangzhou, China
| | - Huhu Zeng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Jiahui Lin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Yongjin Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Nan Yao
- School of Medicine, Jinan University, Guangzhou, China
| | - Tongzheng Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Qi Qi
- School of Medicine, Jinan University, Guangzhou, China
| | - Xiangning Liu
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zhimin Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Minfeng Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China.
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China.
| | - Liangping Xia
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Dongmei Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China.
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China.
| | - Wencai Ye
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China.
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China.
| |
Collapse
|
6
|
Wang S, Huang M, Chen M, Sun Z, Jiao Y, Ye G, Pan J, Ye W, Zhao J, Zhang D. Zoledronic acid and thymosin α1 elicit antitumor immunity against prostate cancer by enhancing tumor inflammation and cytotoxic T cells. J Immunother Cancer 2023; 11:e006381. [PMID: 37295817 PMCID: PMC10277537 DOI: 10.1136/jitc-2022-006381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND Advanced or metastatic prostate cancer (PCa) is still an incurable malignancy with high lethality and a poor prognosis. Despite the remarkable success of immunotherapy against many types of cancer, most patients with PCa receive minimal benefit from current immunotherapeutic strategies, because PCa is an immune cold tumor with scarce T-cell infiltration in the tumor microenvironment. The aim of this study was to develop an effective immunotherapeutic approach for immune cold PCa tumors. METHODS The therapeutic efficacy of androgen deprivation therapy (ADT) and zoledronic acid (ZA) plus thymosin α1 (Tα1) therapy was analyzed retrospectively in patients with advanced or metastatic PCa. The effects and mechanisms by which ZA and Tα1 regulated the immune functions of PCa cells and immune cells were evaluated by a PCa allograft mouse model, flow cytometric analysis, immunohistochemical and immunofluorescence staining assays, and PCR, ELISA, and Western blot analyses. RESULTS In this study, clinical retrospective analysis revealed that ADT combined with ZA plus Tα1 improved the therapeutic outcomes of patients with PCa, which might be associated with an enhanced frequency of T cells. ZA and Tα1 treatment synergistically inhibited the growth of androgen-independent PCa allograft tumors, with increased infiltration of tumor-specific cytotoxic CD8+ T cells and enhanced tumor inflammation. Functionally, ZA and Tα1 treatment relieved immunosuppression in PCa cells, stimulated pro-inflammatory macrophages, and enhanced the cytotoxic function of T cells. Mechanistically, ZA plus Tα1 therapy blocked the MyD88/NF-κB pathway in PCa cells but activated this signaling in macrophages and T cells, altering the tumor immune landscape to suppress PCa progression. CONCLUSIONS These findings uncover a previously undefined role for ZA and Tα1 in inhibiting the disease progression of immune cold PCa tumors by enhancing antitumor immunity and pave the way for the application of ZA plus Tα1 therapy as an immunotherapeutic strategy for treating patients with immunologically unresponsive PCa.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Maohua Huang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Minfeng Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Zhiting Sun
- Research Center of Cancer Diagnosis and Therapy, Department of Oncology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yubo Jiao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Geni Ye
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Jinghua Pan
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Wencai Ye
- Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Jianfu Zhao
- Research Center of Cancer Diagnosis and Therapy, Department of Oncology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Dongmei Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
7
|
Mai W, Liu Q, Li J, Zheng M, Yan F, Liu H, Lei Y, Xu J, Xu J. Comprehensive analysis of the oncogenic and immunological role of FAP and identification of the ceRNA network in human cancers. Aging (Albany NY) 2023; 15:3738-3758. [PMID: 37166418 PMCID: PMC10449273 DOI: 10.18632/aging.204707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/22/2023] [Indexed: 05/12/2023]
Abstract
Fibroblast activation protein-alpha (FAP) is a transmembrane serine protease involving in tissue remodeling. Previous studies report that FAP is highly expressed in certain tumors and participated in oncogenesis. However, there is still lack of systematic and in-depth analysis of FAP based on clinical big data. Here, we comprehensively map the FAP expression profile, prognostic outcome, genetic alteration, immune infiltration across over 30 types of human cancers through multiple datasets including TCGA, CPTAC, and cBioPortal. We find that FAP is up-regulated in most cancer types, and increased FAP expression is associated with advanced pathological stages or poor prognosis in several cancers. Furthermore, FAP is significantly correlated with the infiltration of cancer-associated fibroblasts, macrophages, myeloid dendritic cells, as well as endothelia cells. Immunosuppressive checkpoint proteins or cytokines expression, microsatellite instability and tumor mutational burden analysis also indicate the regulation role of FAP in tumor progression. Gene enrichment analysis demonstrates that ECM-receptor interaction as well as extracellular matrix and structure process are linked to the potential mechanism of FAP in tumor pathogenesis. The ceRNA network is also constructed and identified the involvement of LINC00707/hsa-miR-30e-5p/FAP, LINC02535/hsa-miR-30e-5p/FAP, LINC02535/hsa-miR-30d-5p/FAP, as well as AC026356.1/hsa-miR-30d-5p/FAP axis in tumor progression. In conclusion, our study offers new insights into the oncogenic and immunological role of FAP from a pan-cancer perspective, providing new clues for developing novel targeted anti-tumor strategies.
Collapse
Affiliation(s)
- Weiqian Mai
- School of Medicine, School of Life Science and Engineering, Foshan University, Foshan 528000, China
| | - Qingyou Liu
- School of Medicine, School of Life Science and Engineering, Foshan University, Foshan 528000, China
| | - Jiasheng Li
- School of Medicine, School of Life Science and Engineering, Foshan University, Foshan 528000, China
| | - Mincheng Zheng
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, University Town, Guangzhou 510006, China
| | - Fuman Yan
- Department of Physiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, University Town, Guangzhou 510006, China
| | - Hui Liu
- School of Medicine, School of Life Science and Engineering, Foshan University, Foshan 528000, China
| | - Yuhe Lei
- Department of Pharmacy, Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen 518034, China
| | - Jinwen Xu
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, University Town, Guangzhou 510006, China
- Department of Physiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, University Town, Guangzhou 510006, China
| | - Jiean Xu
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, University Town, Guangzhou 510006, China
- Department of Physiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, University Town, Guangzhou 510006, China
| |
Collapse
|
8
|
Lin K, Huang L, Zhang Y, Chen M, Li Z, Yung KKL, Lv S, Pan Q, Zhang W, Fu J, Li W, Deng Q. The Antiangiogenic and Antitumor Effects of Scoparasin B in Non-Small-Cell Lung Cancer. JOURNAL OF NATURAL PRODUCTS 2023; 86:368-379. [PMID: 36692021 DOI: 10.1021/acs.jnatprod.2c00979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Angiogenesis and vasculogenic mimicry (VM) are crucial for the growth and metastasis of non-small-cell lung cancer (NSCLC). Most tumor angiogenesis inhibitors mainly target endothelial cell-mediated angiogenesis, ignoring tumor-cell-mediated VM and frequently leading to tumor recurrence and metastasis. Thus, development of bioactive molecules interfering with both tumor angiogenesis and VM is necessary. Identifying novel angiogenesis inhibitors from natural products is a promising strategy. Scoparasin B, a pimarane diterpene extracted from a marine-derived fungus, Eutypella sp. F0219, has an antibacterial effect. However, its effect on angiogenesis and VM remains unexplored. In this study, we first certified that scoparasin B showed a strong inhibition effect on angiogenesis and the VM process in vitro and ex vivo. Moreover, scoparasin B prominently impeded tumor growth, angiogenesis, and VM in an NCI-H1299 xenograft model. Further study revealed that scoparasin B restrained tumor angiogenesis and VM by reducing the VEGF-A level and suppressing the VEGF-A/VEGFR2 signaling pathway. This study first demonstrated scoparasin B inhibited tumor angiogenesis, VM, and tumor growth of NSCLC and revealed its underlying mechanism. These new findings further support the potential of scoparasin B as a novel angiogenesis inhibitor and give a hint for further exploring potential angiogenesis inhibitors from natural products.
Collapse
Affiliation(s)
- Kaili Lin
- School of Public Health, Guangzhou Medical University, Guangzhou, 511436, China
- Golden Meditech Center for Neuro Regeneration Sciences, HKBU, Kowloon Tong, Hong Kong 999077, China
| | - Lijuan Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital and The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yu Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital and The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Minshan Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital and The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhan Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital and The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ken Kin Lam Yung
- Department of Biology & Golden Meditech Center for NeuroRegeneration Sciences (GMCNS), Hong Kong Baptist University (HKBU), Kowloon Tong, Hong Kong, China
| | - Sha Lv
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital and The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qianrong Pan
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital and The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Weisong Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital and The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jijun Fu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital and The Sixth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Wanshan Li
- Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, China
| | - Qiudi Deng
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| |
Collapse
|
9
|
Zhao X, Pellow C, Goertz DE. Intravital imaging and cavitation monitoring of antivascular ultrasound in tumor microvasculature. Theranostics 2023; 13:250-266. [PMID: 36593952 PMCID: PMC9800738 DOI: 10.7150/thno.79186] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/09/2022] [Indexed: 12/23/2022] Open
Abstract
Rationale: Focused ultrasound-stimulated microbubbles have been shown to be capable of inducing blood flow shutdown and necrosis in a range of tissue types in an approach termed antivascular ultrasound or nonthermal ablation. In oncology, this approach has demonstrated tumor growth inhibition, and profound synergistic antitumor effects when combined with traditional platforms of chemo-, radiation- and immune-therapies. However, the exposure schemes employed have been broad and underlying mechanisms remain unclear with fundamental questions about exposures, vessel types and sizes involved, and the nature of bubble behaviors and their acoustic emissions resulting in vascular damage - impeding the establishment of standard protocols. Methods: Here, ultrasound transmitters and receivers are integrated into a murine dorsal window chamber tumor model for intravital microscopy studies capable of real-time visual and acoustic monitoring during antivascular ultrasound. Vessel type (normal and tumor-affected), caliber, and viability are assessed under higher pressure conditions (1, 2, and 3 MPa), and cavitation signatures are linked to the biological effects. Results: Vascular events occurred preferentially in tumor-affected vessels with greater incidence in smaller vessels and with more severity as a function of increasing pressure. Vascular blood flow shutdown was found to be due to a combination of focal disruption events and network-related flow changes. Acoustic emissions displayed elevated broadband noise and distinct sub- and ultra-harmonics and their associated third-order peaks with increasing pressure. Conclusions: The observed vascular events taken collectively with identified cavitation signatures provide an improved mechanistic understanding of antivascular ultrasound at the microscale, with implications for establishing a specific treatment protocol and control platform.
Collapse
Affiliation(s)
- Xiaoxiao Zhao
- Department of Medical Biophysics, University of Toronto, Canada
- Sunnybrook Research Institute, Toronto, Canada
| | | | - David E. Goertz
- Department of Medical Biophysics, University of Toronto, Canada
- Sunnybrook Research Institute, Toronto, Canada
| |
Collapse
|
10
|
Xiao M, Shi Y, Jiang S, Cao M, Chen W, Xu Y, Xu Z, Wang K. Recent advances of nanomaterial-based anti-angiogenic therapy in tumor vascular normalization and immunotherapy. Front Oncol 2022; 12:1039378. [PMID: 36523993 PMCID: PMC9745116 DOI: 10.3389/fonc.2022.1039378] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/14/2022] [Indexed: 01/04/2025] Open
Abstract
Anti-angiogenesis therapy and immunotherapy are the first-line therapeutic strategies for various tumor treatments in the clinic, bringing significant advantages for tumor patients. Recent studies have shown that anti-angiogenic therapy can potentiate immunotherapy, with many clinical trials conducted based on the combination of anti-angiogenic agents and immune checkpoint inhibitors (ICIs). However, currently available clinical dosing strategies and tools are limited, emphasizing the need for more improvements. Although significant progress has been achieved, several big questions remained, such as how to achieve cell-specific targeting in the tumor microenvironment? How to improve drug delivery efficiency in tumors? Can nanotechnology be used to potentiate existing clinical drugs and achieve synergistic sensitization effects? Over the recent few years, nanomedicines have shown unique advantages in antitumor research, including cell-specific targeting, improved delivery potentiation, and photothermal effects. Given that the applications of nanomaterials in tumor immunotherapy have been widely reported, this review provides a comprehensive overview of research advances on nanomaterials in anti-angiogenesis therapy, mainly focusing on the immunosuppressive effects of abnormal tumor vessels in the tumor immune microenvironment, the targets and strategies of anti-angiogenesis nanomedicines, and the potential synergistic effects and molecular mechanisms of anti-angiogenic nanomedicines in combination with immunotherapy, ultimately providing new perspectives on the nanomedicine-based synergy between anti-angiogenic and immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Zhiyong Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Kai Wang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| |
Collapse
|
11
|
Bora D, Sharma A, John SE, Shankaraiah N. Development of hydrazide hydrazone-tethered combretastatin-oxindole derivatives as antimitotic agents. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.134675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
12
|
Amini AP, Kirkpatrick JD, Wang CS, Jaeger AM, Su S, Naranjo S, Zhong Q, Cabana CM, Jacks T, Bhatia SN. Multiscale profiling of protease activity in cancer. Nat Commun 2022; 13:5745. [PMID: 36192379 PMCID: PMC9530178 DOI: 10.1038/s41467-022-32988-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 08/24/2022] [Indexed: 11/09/2022] Open
Abstract
Diverse processes in cancer are mediated by enzymes, which most proximally exert their function through their activity. High-fidelity methods to profile enzyme activity are therefore critical to understanding and targeting the pathological roles of enzymes in cancer. Here, we present an integrated set of methods for measuring specific protease activities across scales, and deploy these methods to study treatment response in an autochthonous model of Alk-mutant lung cancer. We leverage multiplexed nanosensors and machine learning to analyze in vivo protease activity dynamics in lung cancer, identifying significant dysregulation that includes enhanced cleavage of a peptide, S1, which rapidly returns to healthy levels with targeted therapy. Through direct on-tissue localization of protease activity, we pinpoint S1 cleavage to the tumor vasculature. To link protease activity to cellular function, we design a high-throughput method to isolate and characterize proteolytically active cells, uncovering a pro-angiogenic phenotype in S1-cleaving cells. These methods provide a framework for functional, multiscale characterization of protease dysregulation in cancer.
Collapse
Affiliation(s)
- Ava P Amini
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Program in Biophysics, Harvard University, Boston, MA, USA
- Microsoft Research New England, Cambridge, MA, USA
| | - Jesse D Kirkpatrick
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Cathy S Wang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alex M Jaeger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Susan Su
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Santiago Naranjo
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Qian Zhong
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Christina M Cabana
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tyler Jacks
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sangeeta N Bhatia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Harvard MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA.
- Wyss Institute at Harvard University, Boston, MA, USA.
- Howard Hughes Medical Institute, Cambridge, MA, USA.
| |
Collapse
|
13
|
Qi M, Fan S, Huang M, Pan J, Li Y, Miao Q, Lyu W, Li X, Deng L, Qiu S, Liu T, Deng W, Chu X, Jiang C, He W, Xia L, Yang Y, Hong J, Qi Q, Yin W, Liu X, Shi C, Chen M, Ye W, Zhang D. Targeting FAPα-expressing hepatic stellate cells overcomes resistance to antiangiogenics in colorectal cancer liver metastasis models. J Clin Invest 2022; 132:e157399. [PMID: 35951441 PMCID: PMC9525122 DOI: 10.1172/jci157399] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
Vessel co-option has been demonstrated to mediate colorectal cancer liver metastasis (CRCLM) resistance to antiangiogenic therapy. The current mechanisms underlying vessel co-option have mainly focused on "hijacker" tumor cells, whereas the function of the "hijackee" sinusoidal blood vessels has not been explored. Here, we found that the occurrence of vessel co-option in bevacizumab-resistant CRCLM xenografts was associated with increased expression of fibroblast activation protein α (FAPα) in the co-opted hepatic stellate cells (HSCs), which was dramatically attenuated in HSC-specific conditional Fap-knockout mice bearing CRCLM allografts. Mechanistically, bevacizumab treatment induced hypoxia to upregulate the expression of fibroblast growth factor-binding protein 1 (FGFBP1) in tumor cells. Gain- or loss-of-function experiments revealed that the bevacizumab-resistant tumor cell-derived FGFBP1 induced FAPα expression by enhancing the paracrine FGF2/FGFR1/ERK1/-2/EGR1 signaling pathway in HSCs. FAPα promoted CXCL5 secretion in HSCs, which activated CXCR2 to promote the epithelial-mesenchymal transition of tumor cells and the recruitment of myeloid-derived suppressor cells. These findings were further validated in tumor tissues derived from patients with CRCLM. Targeting FAPα+ HSCs effectively disrupted the co-opted sinusoidal blood vessels and overcame bevacizumab resistance. Our study highlights the role of FAPα+ HSCs in vessel co-option and provides an effective strategy to overcome the vessel co-option-mediated bevacizumab resistance.
Collapse
Affiliation(s)
- Ming Qi
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Shuran Fan
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Maohua Huang
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Jinghua Pan
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yong Li
- College of Pharmacy, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, and
| | - Qun Miao
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Wenyu Lyu
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Xiaobo Li
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Lijuan Deng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Shenghui Qiu
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Tongzheng Liu
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Weiqing Deng
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Xiaodong Chu
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Chang Jiang
- Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wenzhuo He
- Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Liangping Xia
- Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yunlong Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jian Hong
- School of Medicine, Jinan University, Guangzhou, China
| | - Qi Qi
- School of Medicine, Jinan University, Guangzhou, China
| | - Wenqian Yin
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Xiangning Liu
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Changzheng Shi
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Minfeng Chen
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Wencai Ye
- College of Pharmacy, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, and
| | - Dongmei Zhang
- College of Pharmacy, Jinan University, Guangzhou, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, and
| |
Collapse
|
14
|
Huang M, Lin Y, Wang C, Deng L, Chen M, Assaraf YG, Chen ZS, Ye W, Zhang D. New insights into antiangiogenic therapy resistance in cancer: Mechanisms and therapeutic aspects. Drug Resist Updat 2022; 64:100849. [PMID: 35842983 DOI: 10.1016/j.drup.2022.100849] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Angiogenesis is a hallmark of cancer and is required for tumor growth and progression. Antiangiogenic therapy has been revolutionarily developing and was approved for the treatment of various types of cancer for nearly two decades, among which bevacizumab and sorafenib continue to be the two most frequently used antiangiogenic drugs. Although antiangiogenic therapy has brought substantial survival benefits to many cancer patients, resistance to antiangiogenic drugs frequently occurs during clinical treatment, leading to poor outcomes and treatment failure. Cumulative evidence has demonstrated that the intricate interplay among tumor cells, bone marrow-derived cells, and local stromal cells critically allows for tumor escape from antiangiogenic therapy. Currently, drug resistance has become the main challenge that hinders the therapeutic efficacies of antiangiogenic therapy. In this review, we describe and summarize the cellular and molecular mechanisms conferring tumor drug resistance to antiangiogenic therapy, which was predominantly associated with redundancy in angiogenic signaling molecules (e.g., VEGFs, GM-CSF, G-CSF, and IL17), alterations in biological processes of tumor cells (e.g., tumor invasiveness and metastasis, stemness, autophagy, metabolic reprogramming, vessel co-option, and vasculogenic mimicry), increased recruitment of bone marrow-derived cells (e.g., myeloid-derived suppressive cells, tumor-associated macrophages, and tumor-associated neutrophils), and changes in the biological functions and features of local stromal cells (e.g., pericytes, cancer-associated fibroblasts, and endothelial cells). We also review potential biomarkers to predict the response to antiangiogenic therapy in cancer patients, which mainly consist of imaging biomarkers, cellular and extracellular proteins, a certain type of bone marrow-derived cells, local stromal cell content (e.g., pericyte coverage) as well as serum or plasma biomarkers (e.g., non-coding RNAs). Finally, we highlight the recent advances in combination strategies with the aim of enhancing the response to antiangiogenic therapy in cancer patients and mouse models. This review introduces a comprehensive understanding of the mechanisms and biomarkers associated with the evasion of antiangiogenic therapy in cancer, providing an outlook for developing more effective approaches to promote the therapeutic efficacy of antiangiogenic therapy.
Collapse
Affiliation(s)
- Maohua Huang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Yuning Lin
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Chenran Wang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Lijuan Deng
- Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Minfeng Chen
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Institute for Biotechnology, St. John's University, NY 11439, USA.
| | - Wencai Ye
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| | - Dongmei Zhang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
15
|
Fu J, Zeng W, Chen M, Huang L, Li S, Li Z, Pan Q, Lv S, Yang X, Wang Y, Yi M, Zhang J, Lei X. Apigenin suppresses tumor angiogenesis and growth via inhibiting HIF-1α expression in non-small cell lung carcinoma. Chem Biol Interact 2022; 361:109966. [PMID: 35513012 DOI: 10.1016/j.cbi.2022.109966] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/12/2022] [Accepted: 04/25/2022] [Indexed: 12/25/2022]
Abstract
Tumor angiogenesis inhibitors such as Bevacizumab, Ramucirumab and Endostar have been applied to the therapy of non-small cell lung carcinoma (NSCLC) patients, especially for lung adenocarcinoma (LUAD). However, several safe concerns such as neutropenia, febrile neutropenia and hypertension pulmonary hemorrhage limit their further development. And they often showed poor efficacy and serious side effect for lung squamous cell carcinoma (LUSC) patient. Thus, identification of effective and safe tumor angiogenesis inhibitor for NSCLC therapy is warranted. Apigenin is a bioflavonoid with potential anti-tumor effect and perfect safety, but its effect on tumor angiogenesis and underlying mechanism are still unclear. Herein, we found that apigenin not merely suppressed endothelial cells related motilities but also reduced pericyte coverage. Further research showed that apigenin had strong suppressive activity against HIF-1α expression and its downstream VEGF-A/VEGFR2 and PDGF-BB/PDGFβR signaling pathway. Apigenin also reduced microvessel density and pericyte coverage on the xengraft model of NCI-H1299 cells, leading to suppression of tumor growth. Moreover, apigenein showed perfect anti-angiogenic effect in xengraft model of LUSC cell NCI-H1703 cells, indicating it may be developed into a potential angiogenesis inhibitor for LUSC patient. Collectively, our study provides new insights into the anti-tumor mechanism of apigenin and suggests that apigenin is a safe and effective angiogenesis inhibitor for NSCLC therapy.
Collapse
Affiliation(s)
- Jijun Fu
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Wenjuan Zeng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, No. 613, Huangpu Road, Guangzhou, China
| | - Minshan Chen
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Lijuan Huang
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Songpei Li
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Zhan Li
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Qianrong Pan
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Sha Lv
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Xiangyu Yang
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Ying Wang
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Mengmeng Yi
- Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, China.
| | - Jianye Zhang
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China.
| | - Xueping Lei
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, PR China.
| |
Collapse
|
16
|
Wang Y, Sun Q, Ye Y, Sun X, Xie S, Zhan Y, Song J, Fan X, Zhang B, Yang M, Lv L, Hosaka K, Yang Y, Nie G. FGF-2 signaling in nasopharyngeal carcinoma modulates pericyte-macrophage crosstalk and metastasis. JCI Insight 2022; 7:157874. [PMID: 35439170 PMCID: PMC9220856 DOI: 10.1172/jci.insight.157874] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/15/2022] [Indexed: 12/24/2022] Open
Abstract
Molecular signaling in the tumor microenvironment (TME) is complex, and crosstalks among various cell compartments in supporting metastasis remain poorly understood. In particular, the role of vascular pericytes, a critical cellular component in the TME, in cancer invasion and metastasis warrants further investigation. Here we report an elevation of FGF-2 signaling in both nasopharyngeal carcinoma (NPC) patient samples and xenograft mouse models promotes NPC metastasis. Mechanistically, tumor cell-derived FGF-2 strongly promoted pericyte proliferation and pericyte-specific expression of an orphan chemokine (C-X-C motif) ligand 14 (CXCL14) via FGFR1- AHR signaling. Gain and loss-of-function experiments validated that pericyte-derived CXCL14 promoted macrophage recruitment and polarization towards an M2-like phenotype. Genetic knockdown of FGF2 or genetic depletion of tumoral pericytes blocked CXCL14 expression and tumor-associated macrophage (TAM) infiltration. Pharmacological inhibition of TAMs by clodronate liposomes treatment resulted in a reduction of FGF-2-induced pulmonary metastasis. Together, these findings shed light on the inflammatory role of tumoral pericytes in promoting TAM-mediated metastasis. We provide mechanistic insight into an FGF-2-FGFR1-pericyte-CXCL14-TAM stromal communication axis in NPC and propose an effective anti-metastasis therapy concept by targeting a pericyte-derived inflammation for NPC or FGF-2-high tumors.
Collapse
Affiliation(s)
- Yujie Wang
- Department of Otolaryngology, Shenzhen Second People's Hospital, Shenzhen, China
| | - Qi Sun
- Fudan University, Shanghai, China
| | - Ying Ye
- Department of Oral Implantology, Tongji University, Shanghai, China
| | - Xiaoting Sun
- Department of Cellular and Genetic Medicine, Fudan University, Shanghai, China
| | - Sisi Xie
- Department of Cellular and Genetic Medicine, Fudan University, Shanghai, China
| | - Yuhang Zhan
- Department of Cellular and Genetic Medicine, Fudan University, Shanghai, China
| | - Jian Song
- Department of Otolaryngology, Shenzhen Second People's Hospital, Shenzhen, China
| | - Xiaoqin Fan
- Department of Otolaryngology, Shenzhen Second People's Hospital, Shenzhen, China
| | - Bin Zhang
- Department of Otolaryngology, Shenzhen Second People's Hospital, Shenzhen, China
| | - Ming Yang
- Department of Otolaryngology, Shenzhen Second People's Hospital, Shenzhen, China
| | - Lei Lv
- Department of Biochemistry and Molecular Biology, Fudan University, Shanghai, China
| | - Kayoko Hosaka
- Department of Microbiology, Karolinska Institute, Stockholm, Sweden
| | - Yunlong Yang
- Department of Cellular and Genetic Medicine, Fudan University, Shanghai, China
| | - Guohui Nie
- Department of Otolaryngology, Shenzhen Second People's Hospital, Shenzhen, China
| |
Collapse
|
17
|
Current Nano-Strategies to Target Tumor Microenvironment to Improve Anti-Tumor efficiency. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
18
|
Ye G, Huang M, Li Y, Ouyang J, Chen M, Wen Q, Li X, Zeng H, Long P, Fan Z, Yin J, Ye W, Zhang D. The FAP α -activated prodrug Z-GP-DAVLBH inhibits the growth and pulmonary metastasis of osteosarcoma cells by suppressing the AXL pathway. Acta Pharm Sin B 2022; 12:1288-1304. [PMID: 35530139 PMCID: PMC9072247 DOI: 10.1016/j.apsb.2021.08.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/27/2021] [Accepted: 07/30/2021] [Indexed: 01/02/2023] Open
Abstract
Osteosarcoma is a kind of bone tumor with highly proliferative and invasive properties, a high incidence of pulmonary metastasis and a poor prognosis. Chemotherapy is the mainstay of treatment for osteosarcoma. Currently, there are no molecular targeted drugs approved for osteosarcoma treatment, particularly effective drugs for osteosarcoma with pulmonary metastases. It has been reported that fibroblast activation protein alpha (FAPα) is upregulated in osteosarcoma and critically associated with osteosarcoma progression and metastasis, demonstrating that FAPα-targeted agents might be a promising therapeutic strategy for osteosarcoma. In the present study, we reported that the FAPα-activated vinblastine prodrug Z-GP-DAVLBH exhibited potent antitumor activities against FAPα-positive osteosarcoma cells in vitro and in vivo. Z-GP-DAVLBH inhibited the growth and induced the apoptosis of osteosarcoma cells. Importantly, it also decreased the migration and invasion capacities and reversed epithelial–mesenchymal transition (EMT) of osteosarcoma cells in vitro and suppressed pulmonary metastasis of osteosarcoma xenografts in vivo. Mechanistically, Z-GP-DAVLBH suppressed the AXL/AKT/GSK-3β/β-catenin pathway, leading to inhibition of the growth and metastatic spread of osteosarcoma cells. These findings demonstrate that Z-GP-DAVLBH is a promising agent for the treatment of FAPα-positive osteosarcoma, particularly osteosarcoma with pulmonary metastases.
Collapse
|
19
|
Synthesis and anticancer activity of Boc-Gly-Pro dipeptide-annonaceous acetogenin prodrugs targeting fibroblast activation protein or other hydrolytic enzymes. Med Chem Res 2022. [DOI: 10.1007/s00044-022-02857-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
20
|
Andrea AE, Chiron A, Mallah S, Bessoles S, Sarrabayrouse G, Hacein-Bey-Abina S. Advances in CAR-T Cell Genetic Engineering Strategies to Overcome Hurdles in Solid Tumors Treatment. Front Immunol 2022; 13:830292. [PMID: 35211124 PMCID: PMC8861853 DOI: 10.3389/fimmu.2022.830292] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/18/2022] [Indexed: 12/15/2022] Open
Abstract
During this last decade, adoptive transfer of T lymphocytes genetically modified to express chimeric antigen receptors (CARs) emerged as a valuable therapeutic strategy in hematological cancers. However, this immunotherapy has demonstrated limited efficacy in solid tumors. The main obstacle encountered by CAR-T cells in solid malignancies is the immunosuppressive tumor microenvironment (TME). The TME impedes tumor trafficking and penetration of T lymphocytes and installs an immunosuppressive milieu by producing suppressive soluble factors and by overexpressing negative immune checkpoints. In order to overcome these hurdles, new CAR-T cells engineering strategies were designed, to potentiate tumor recognition and infiltration and anti-cancer activity in the hostile TME. In this review, we provide an overview of the major mechanisms used by tumor cells to evade immune defenses and we critically expose the most optimistic engineering strategies to make CAR-T cell therapy a solid option for solid tumors.
Collapse
Affiliation(s)
- Alain E. Andrea
- Laboratoire de Biochimie et Thérapies Moléculaires, Faculté de Pharmacie, Université Saint Joseph de Beyrouth, Beirut, Lebanon
| | - Andrada Chiron
- Université de Paris, CNRS, INSERM, UTCBS, Unité des technologies Chimiques et Biologiques pour la Santé, Paris, France
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, Hôpital Kremlin-Bicêtre, Assistance Publique-Hôpitaux de Paris, Le-Kremlin-Bicêtre, France
| | - Sarah Mallah
- Faculty of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Stéphanie Bessoles
- Université de Paris, CNRS, INSERM, UTCBS, Unité des technologies Chimiques et Biologiques pour la Santé, Paris, France
| | - Guillaume Sarrabayrouse
- Université de Paris, CNRS, INSERM, UTCBS, Unité des technologies Chimiques et Biologiques pour la Santé, Paris, France
| | - Salima Hacein-Bey-Abina
- Université de Paris, CNRS, INSERM, UTCBS, Unité des technologies Chimiques et Biologiques pour la Santé, Paris, France
- Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, Hôpital Kremlin-Bicêtre, Assistance Publique-Hôpitaux de Paris, Le-Kremlin-Bicêtre, France
| |
Collapse
|
21
|
Clinical summary of fibroblast activation protein inhibitor-based radiopharmaceuticals: cancer and beyond. Eur J Nucl Med Mol Imaging 2022; 49:2844-2868. [DOI: 10.1007/s00259-022-05706-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/23/2022] [Indexed: 02/06/2023]
|
22
|
Verhulst E, Garnier D, De Meester I, Bauvois B. Validating Cell Surface Proteases as Drug Targets for Cancer Therapy: What Do We Know, and Where Do We Go? Cancers (Basel) 2022; 14:624. [PMID: 35158891 PMCID: PMC8833564 DOI: 10.3390/cancers14030624] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
Cell surface proteases (also known as ectoproteases) are transmembrane and membrane-bound enzymes involved in various physiological and pathological processes. Several members, most notably dipeptidyl peptidase 4 (DPP4/CD26) and its related family member fibroblast activation protein (FAP), aminopeptidase N (APN/CD13), a disintegrin and metalloprotease 17 (ADAM17/TACE), and matrix metalloproteinases (MMPs) MMP2 and MMP9, are often overexpressed in cancers and have been associated with tumour dysfunction. With multifaceted actions, these ectoproteases have been validated as therapeutic targets for cancer. Numerous inhibitors have been developed to target these enzymes, attempting to control their enzymatic activity. Even though clinical trials with these compounds did not show the expected results in most cases, the field of ectoprotease inhibitors is growing. This review summarizes the current knowledge on this subject and highlights the recent development of more effective and selective drugs targeting ectoproteases among which small molecular weight inhibitors, peptide conjugates, prodrugs, or monoclonal antibodies (mAbs) and derivatives. These promising avenues have the potential to deliver novel therapeutic strategies in the treatment of cancers.
Collapse
Affiliation(s)
- Emile Verhulst
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, 2000 Antwerp, Belgium; (E.V.); (I.D.M.)
| | - Delphine Garnier
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, F-75006 Paris, France;
| | - Ingrid De Meester
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, 2000 Antwerp, Belgium; (E.V.); (I.D.M.)
| | - Brigitte Bauvois
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, F-75006 Paris, France;
| |
Collapse
|
23
|
Tumor perivascular cell-derived extracellular vesicles promote angiogenesis via the Gas6/Axl pathway. Cancer Lett 2022; 524:131-143. [PMID: 34678434 DOI: 10.1016/j.canlet.2021.10.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/29/2021] [Accepted: 10/17/2021] [Indexed: 12/19/2022]
Abstract
Aberrant angiogenesis is a hallmark of cancer and is critically associated with tumor progression. Perivascular cells are essential components of blood vessels, and the role of tumor perivascular cell-derived extracellular vesicles (TPC-EVs) in angiogenesis remains elusive. In the present study, using genetic mouse models and pharmacological inhibitors, we found that ablation of perivascular cells inhibited angiogenesis in allografted colorectal cancer tumors. Further studies demonstrated that TPC-EVs promoted the proliferation, migration, invasion, viability, and tube formation of HUVECs. They also facilitated vessel spouting in rat aortic rings and induced neovascularization in chick chorioallantoic membranes (CAMs). Silencing of Gas6 or blockade of the Axl pathway suppressed TPC-EV-induced angiogenesis in vitro and ex vivo. Moreover, inhibition of the Gas6/Axl signaling pathway impaired TPC-EV-mediated angiogenesis in vivo. Our findings present a deeper insight into the biological functions of TPCs and TPC-EVs in tumor angiogenesis and demonstrate that TPC-EV-derived Gas6 could be an attractive and innovative regulator of tumor angiogenesis.
Collapse
|
24
|
Kochetkova M, Samuel MS. Differentiation of the tumor microenvironment: are CAFs the Organizer? Trends Cell Biol 2021; 32:285-294. [PMID: 34895986 DOI: 10.1016/j.tcb.2021.11.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 12/13/2022]
Abstract
Cancers contain a suite of genetically stable cells within an extracellular matrix, collectively termed the tumor microenvironment (TME). The TME strongly influences disease outcome for patients. Gleaning clues from the literature, we propose that the TME should be viewed not as disparate populations of cells constituting a pathological lesion, but as a cohesive tissue constituting a novel pathological organ, arising from the coordinated differentiation of its constituent cell types - a process we have termed tumor-associated neodifferentiation (TAND). We also discuss why cancer-associated fibroblasts (CAFs) may assume the role of Organizer of this organ, directing the recruitment and differentiation of cells within the TME. Viewing the microenvironment in this way will reveal new cancer vulnerabilities that may be exploited for therapy.
Collapse
Affiliation(s)
- Marina Kochetkova
- Centre for Cancer Biology, an alliance between SA Pathology and the University of South Australia, Adelaide, SA 5000, Australia
| | - Michael Susithiran Samuel
- Centre for Cancer Biology, an alliance between SA Pathology and the University of South Australia, Adelaide, SA 5000, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia.
| |
Collapse
|
25
|
Yu X, Su Q, Chang X, Chen K, Yuan P, Liu T, Tian R, Bai Y, Zhang Y, Chen X. Multimodal obstruction of tumorigenic energy supply via bionic nanocarriers for effective tumor therapy. Biomaterials 2021; 278:121181. [PMID: 34653932 DOI: 10.1016/j.biomaterials.2021.121181] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 09/05/2021] [Accepted: 10/07/2021] [Indexed: 12/11/2022]
Abstract
Sufficient energy generation based on effective transport of nutrient via abundant blood vessels in tumor tissue and subsequent oxidative metabolism in mitochondria is critical for growth, proliferation and migration of tumor. Thus the strategy to cut off this transport pathway (blood vessels) and simultaneously close the power house (mitochondria) is highly desired for tumor treatment. Herein, we fabricated a bionic nanocarrier with core-shell-corona structure to give selective and effective tumor therapy via stepwise destruction of existed tumor vessel, inhibition of tumor angiogenesis and dysfunction of tumor mitochondria. The core of this bionic nanocarrier consists of combretastatin A4 phosphate (CA4P) and vitamin K2 (VK2) co-loaded mesoporous silica nanoparticle (MSNs), which is in charge of the vasculature destruction and mitochondrial dysfunction after cargos release. The N-tert-butylacrylamide (TBAM) and tri-sulfated N-acetylglucosamine (TSAG) shell served as artificial affinity reagent against vascular endothelial growth factor (VEGF) for angiogenesis inhibition. As to guarantee that these actions only happened in tumor, the hyaluronic acid (HA) corona was introduced to endow the nanocarrier with tumor targeting property and stimuli-responsiveness for accurate therapy. Both in vitro and in vivo results indicated that the CA4P/VK2-MSNs-TBAM/TSAG-HA (CVMMGH for short) nanocarrier combined well-controllable manipulation of tumor vasculature and tumor mitochondria to effectivly cut off the tumorigenic energy supply, which performed significant inhibition of tumor growth, demonstrating the great candidate of our strategy for effective tumor therapy.
Collapse
Affiliation(s)
- Xiaoqian Yu
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Qi Su
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Xiaowei Chang
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Kun Chen
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China
| | - Pingyun Yuan
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Tao Liu
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Ran Tian
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yongkang Bai
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, PR China.
| | - Xin Chen
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| |
Collapse
|
26
|
Juillerat-Jeanneret L, Tafelmeyer P, Golshayan D. Regulation of Fibroblast Activation Protein-α Expression: Focus on Intracellular Protein Interactions. J Med Chem 2021; 64:14028-14045. [PMID: 34523930 DOI: 10.1021/acs.jmedchem.1c01010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The prolyl-specific peptidase fibroblast activation protein-α (FAP-α) is expressed at very low or undetectable levels in nondiseased human tissues but is selectively induced in activated (myo)fibroblasts at sites of tissue remodeling in fibrogenic processes. In normal regenerative processes involving transient fibrosis FAP-α+(myo)fibroblasts disappear from injured tissues, replaced by cells with a normal FAP-α- phenotype. In chronic uncontrolled pathological fibrosis FAP-α+(myo)fibroblasts permanently replace normal tissues. The mechanisms of regulation and elimination of FAP-α expression in(myo)fibroblasts are unknown. According to a yeast two-hybrid screen and protein databanks search, we propose that the intracellular (co)-chaperone BAG6/BAT3 can interact with FAP-α, mediated by the BAG6/BAT3 Pro-rich domain, inducing proteosomal degradation of FAP-α protein under tissue homeostasis. In this Perspective, we discuss our findings in the context of current knowledge on the regulation of FAP-α expression and comment potential therapeutic strategies for uncontrolled fibrosis, including small molecule degraders (PROTACs)-modified FAP-α targeted inhibitors.
Collapse
Affiliation(s)
- Lucienne Juillerat-Jeanneret
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), CH1011 Lausanne, Switzerland.,University Institute of Pathology, CHUV and UNIL, CH1011 Lausanne, Switzerland
| | - Petra Tafelmeyer
- Hybrigenics Services, Laboratories and Headquarters-Paris, 1 rue Pierre Fontaine, 91000 Evry, France.,Hybrigenics Corporation, Cambridge Innovation Center, 50 Milk Street, Cambridge, Massachusetts 02142, United States
| | - Dela Golshayan
- Transplantation Center and Transplantation Immunopathology Laboratory, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), CH1011 Lausanne, Switzerland
| |
Collapse
|
27
|
Guo Y, Wang H, Gerberich JL, Odutola SO, Charlton-Sevcik AK, Li M, Tanpure RP, Tidmore JK, Trawick ML, Pinney KG, Mason RP, Liu L. Imaging-Guided Evaluation of the Novel Small-Molecule Benzosuberene Tubulin-Binding Agent KGP265 as a Potential Therapeutic Agent for Cancer Treatment. Cancers (Basel) 2021; 13:cancers13194769. [PMID: 34638255 PMCID: PMC8507561 DOI: 10.3390/cancers13194769] [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: 08/03/2021] [Revised: 09/10/2021] [Accepted: 09/17/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Vascular-disrupting agents promise significant therapeutic efficacy against solid tumors by selectively damaging tumor-associated vasculature. Dynamic BLI and oxygen-enhanced multispectral optoacoustic tomography (OE-MSOT) were used to compare vascular shutdown following administration of KGP265. BLI signal and vascular oxygenation response (ΔsO2) to a gas breathing challenge were both significantly reduced within 2 h indicating vascular disruption, which continued over 24 h. Twice-weekly doses of KGP265 caused a significant growth delay in MDA-MB-231 human breast tumor xenografts and 4T1 syngeneic breast tumors growing orthotopically in mice. Abstract The selective disruption of tumor-associated vasculature represents an attractive therapeutic approach. We have undertaken the first in vivo evaluation of KGP265, a water-soluble prodrug of a benzosuberene-based tubulin-binding agent, and found promising vascular-disrupting activity in three distinct tumor types. Dose escalation in orthotopic MDA-MB-231-luc breast tumor xenografts in mice indicated that higher doses produced more effective vascular shutdown, as revealed by dynamic bioluminescence imaging (BLI). In syngeneic orthotopic 4T1-luc breast and RENCA-luc kidney tumors, dynamic BLI and oxygen enhanced multispectral optoacoustic tomography (OE-MSOT) were used to compare vascular shutdown following the administration of KGP265 (7.5 mg/kg). The BLI signal and vascular oxygenation response (ΔsO2) to a gas breathing challenge were both significantly reduced within 2 h, indicating vascular disruption, which continued over 24 h. A correlative histology confirmed increased necrosis and hemorrhage. Twice-weekly doses of KGP265 caused significant growth delay in both MDA-MB-231 and 4T1 breast tumors, with no obvious systemic toxicity. A combination with carboplatin produced significantly greater tumor growth delay than carboplatin alone, though significant carboplatin-associated toxicity was observed (whole-body weight loss). KGP265 was found to be effective at low concentrations, generating long-term vascular shutdown and tumor growth delay, thus providing strong rationale for further development, particularly in combination therapies.
Collapse
Affiliation(s)
- Yihang Guo
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (Y.G.); (H.W.); (J.L.G.); (M.L.)
- Department of Gastrointestinal Surgery, The Third XiangYa Hospital of Central South University, Changsha 410013, China
| | - Honghong Wang
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (Y.G.); (H.W.); (J.L.G.); (M.L.)
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jeni L. Gerberich
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (Y.G.); (H.W.); (J.L.G.); (M.L.)
| | - Samuel O. Odutola
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (S.O.O.); (A.K.C.-S.); (R.P.T.); (J.K.T.); (M.L.T.); (K.G.P.)
| | - Amanda K. Charlton-Sevcik
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (S.O.O.); (A.K.C.-S.); (R.P.T.); (J.K.T.); (M.L.T.); (K.G.P.)
| | - Maoping Li
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (Y.G.); (H.W.); (J.L.G.); (M.L.)
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Rajendra P. Tanpure
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (S.O.O.); (A.K.C.-S.); (R.P.T.); (J.K.T.); (M.L.T.); (K.G.P.)
| | - Justin K. Tidmore
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (S.O.O.); (A.K.C.-S.); (R.P.T.); (J.K.T.); (M.L.T.); (K.G.P.)
| | - Mary Lynn Trawick
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (S.O.O.); (A.K.C.-S.); (R.P.T.); (J.K.T.); (M.L.T.); (K.G.P.)
| | - Kevin G. Pinney
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (S.O.O.); (A.K.C.-S.); (R.P.T.); (J.K.T.); (M.L.T.); (K.G.P.)
| | - Ralph P. Mason
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (Y.G.); (H.W.); (J.L.G.); (M.L.)
- Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Correspondence: (R.P.M.); (L.L.)
| | - Li Liu
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (Y.G.); (H.W.); (J.L.G.); (M.L.)
- Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Correspondence: (R.P.M.); (L.L.)
| |
Collapse
|
28
|
Xin L, Gao J, Zheng Z, Chen Y, Lv S, Zhao Z, Yu C, Yang X, Zhang R. Fibroblast Activation Protein-α as a Target in the Bench-to-Bedside Diagnosis and Treatment of Tumors: A Narrative Review. Front Oncol 2021; 11:648187. [PMID: 34490078 PMCID: PMC8416977 DOI: 10.3389/fonc.2021.648187] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
Fibroblast activation protein-α (FAP) is a type II integral serine protease that is specifically expressed by activated fibroblasts. Cancer-associated fibroblasts (CAFs) in the tumor stroma have an abundant and stable expression of FAP, which plays an important role in promoting tumor growth, invasion, metastasis, and immunosuppression. For example, in females with a high incidence of breast cancer, CAFs account for 50–70% of the cells in the tumor’s microenvironment. CAF overexpression of FAP promotes tumor development and metastasis by influencing extracellular matrix remodeling, intracellular signaling, angiogenesis, epithelial-to-mesenchymal transition, and immunosuppression. This review discusses the basic biological characteristics of FAP and its applications in the diagnosis and treatment of various cancers. We review the emerging basic and clinical research data regarding the use of nanomaterials that target FAP.
Collapse
Affiliation(s)
- Lei Xin
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Jinfang Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Ziliang Zheng
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Yiyou Chen
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Shuxin Lv
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Zhikai Zhao
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Chunhai Yu
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaotang Yang
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| | - Ruiping Zhang
- Department of Radiology, Affiliated Tumor Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
29
|
Lees DM, Reynolds LE, Pedrosa AR, Roy-Luzarraga M, Hodivala-Dilke KM. Phosphorylation of pericyte FAK-Y861 affects tumour cell apoptosis and tumour blood vessel regression. Angiogenesis 2021; 24:471-482. [PMID: 33730293 PMCID: PMC8292267 DOI: 10.1007/s10456-021-09776-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 02/25/2021] [Indexed: 12/20/2022]
Abstract
Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase that is overexpressed in many cancer types and in vivo studies have shown that vascular endothelial cell FAK expression and FAK-phosphorylation at tyrosine (Y) 397, and subsequently FAK-Y861, are important in tumour angiogenesis. Pericytes also play a vital role in regulating tumour blood vessel stabilisation, but the specific involvement of pericyte FAK-Y397 and FAK-Y861 phosphorylation in tumour blood vessels is unknown. Using PdgfrβCre + ;FAKWT/WT, PdgfrβCre + ;FAKY397F/Y397F and PdgfrβCre + ;FAKY861F/Y861F mice, our data demonstrate that Lewis lung carcinoma tumour growth, tumour blood vessel density, blood vessel perfusion and pericyte coverage were affected only in late stage tumours in PdgfrβCre + ;FAKY861F/Y861F but not PdgfrβCre + ;FAKY397F/Y397F mice. Further examination indicates a dual role for pericyte FAK-Y861 phosphorylation in the regulation of tumour vessel regression and also in the control of pericyte derived signals that influence apoptosis in cancer cells. Overall this study identifies the role of pericyte FAK-Y861 in the regulation of tumour vessel regression and tumour growth control and that non-phosphorylatable FAK-Y861F in pericytes reduces tumour growth and blood vessel density.
Collapse
Affiliation(s)
- Delphine M Lees
- Adhesion and Angiogenesis Laboratory, Centre for Tumour Microenvironment, Barts Cancer Institute - a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Louise E Reynolds
- Adhesion and Angiogenesis Laboratory, Centre for Tumour Microenvironment, Barts Cancer Institute - a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Ana Rita Pedrosa
- Adhesion and Angiogenesis Laboratory, Centre for Tumour Microenvironment, Barts Cancer Institute - a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Marina Roy-Luzarraga
- Adhesion and Angiogenesis Laboratory, Centre for Tumour Microenvironment, Barts Cancer Institute - a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Kairbaan M Hodivala-Dilke
- Adhesion and Angiogenesis Laboratory, Centre for Tumour Microenvironment, Barts Cancer Institute - a CR-UK Centre of Excellence, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK.
| |
Collapse
|
30
|
Sun R, Kong X, Qiu X, Huang C, Wong PP. The Emerging Roles of Pericytes in Modulating Tumor Microenvironment. Front Cell Dev Biol 2021; 9:676342. [PMID: 34179005 PMCID: PMC8232225 DOI: 10.3389/fcell.2021.676342] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/06/2021] [Indexed: 12/17/2022] Open
Abstract
Pericytes (PCs), known as mural cells, play an important blood vessel (BV) supporting role in regulating vascular stabilization, permeability and blood flow in microcirculation as well as blood brain barrier. In carcinogenesis, defective interaction between PCs and endothelial cells (ECs) contributes to the formation of leaky, chaotic and dysfunctional vasculature in tumors. However, recent works from other laboratories and our own demonstrate that the direct interaction between PCs and other stromal cells/cancer cells can modulate tumor microenvironment (TME) to favor cancer growth and progression, independent of its BV supporting role. Furthermore, accumulating evidence suggests that PCs have an immunomodulatory role. In the current review, we focus on recent advancement in understanding PC's regulatory role in the TME by communicating with ECs, immune cells, and tumor cells, and discuss how we can target PC's functions to re-model TME for an improved cancer treatment strategy.
Collapse
Affiliation(s)
- Ruipu Sun
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiangzhan Kong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyi Qiu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Cheng Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ping-Pui Wong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
31
|
Huang M, Chen M, Qi M, Ye G, Pan J, Shi C, Yang Y, Zhao L, Mo X, Zhang Y, Li Y, Zhong J, Lu W, Li X, Zhang J, Lin J, Luo L, Liu T, Tang PMK, Hong A, Cao Y, Ye W, Zhang D. Perivascular cell-derived extracellular vesicles stimulate colorectal cancer revascularization after withdrawal of antiangiogenic drugs. J Extracell Vesicles 2021; 10:e12096. [PMID: 34035882 PMCID: PMC8138700 DOI: 10.1002/jev2.12096] [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: 12/02/2020] [Revised: 04/02/2021] [Accepted: 05/04/2021] [Indexed: 12/22/2022] Open
Abstract
Antiangiogenic tyrosine kinase inhibitors (AA‐TKIs) have become a promising therapeutic strategy for colorectal cancer (CRC). In clinical practice, a significant proportion of cancer patients temporarily discontinue AA‐TKI treatment due to recurrent toxicities, economic burden or acquired resistance. However, AA‐TKI therapy withdrawal‐induced tumour revascularization frequently occurs, hampering the clinical application of AA‐TKIs. Here, this study demonstrates that tumour perivascular cells mediate tumour revascularization after withdrawal of AA‐TKI therapy. Pharmacological inhibition and genetic ablation of perivascular cells largely attenuate the rebound effect of CRC vascularization in the AA‐TKI cessation experimental settings. Mechanistically, tumour perivascular cell‐derived extracellular vehicles (TPC‐EVs) contain Gas6 that instigates the recruitment of endothelial progenitor cells (EPCs) for tumour revascularization via activating the Axl pathway. Gas6 silence and an Axl inhibitor markedly inhibit tumour revascularization by impairing EPC recruitment. Consequently, combination therapy of regorafenib with the Axl inhibitor improves overall survival in mice metastatic CRC model by inhibiting tumour growth. Together, these data shed new mechanistic insights into perivascular cells in off‐AA‐TKI‐induced tumour revascularization and indicate that blocking the Axl signalling may provide an attractive anticancer approach for sustaining long‐lasting angiostatic effects to improve the therapeutic outcomes of antiangiogenic drugs in CRC.
Collapse
Affiliation(s)
- Maohua Huang
- College of Pharmacy Jinan University Guangzhou China
| | - Minfeng Chen
- College of Pharmacy Jinan University Guangzhou China
| | - Ming Qi
- College of Pharmacy Jinan University Guangzhou China
| | - Geni Ye
- College of Pharmacy Jinan University Guangzhou China
| | - Jinghua Pan
- Department of General Surgery the First Affiliated Hospital of Jinan University Guangzhou China
| | - Changzheng Shi
- Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation the First Affiliated Hospital of Jinan University Guangzhou China
| | - Yunlong Yang
- Department of Cellular and Genetic Medicine School of Basic Medical Sciences Fudan University Shanghai China
| | - Luyu Zhao
- Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation the First Affiliated Hospital of Jinan University Guangzhou China
| | - Xukai Mo
- Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation the First Affiliated Hospital of Jinan University Guangzhou China
| | - Yiran Zhang
- Department of General Surgery the First Affiliated Hospital of Jinan University Guangzhou China
| | - Yong Li
- College of Pharmacy Jinan University Guangzhou China
| | | | - Weijin Lu
- College of Pharmacy Jinan University Guangzhou China
| | - Xiaobo Li
- College of Pharmacy Jinan University Guangzhou China
| | - Jiayan Zhang
- College of Pharmacy Jinan University Guangzhou China
| | - Jinrong Lin
- Department of Obstetrics the First Affiliated Hospital of Jinan University Guangzhou China
| | - Liangping Luo
- Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation the First Affiliated Hospital of Jinan University Guangzhou China
| | - Tongzheng Liu
- College of Pharmacy Jinan University Guangzhou China
| | - Patrick Ming-Kuen Tang
- Department of Anatomical and Cellular Pathology Prince of Wales Hospital The Chinese University of Hong Kong Sha Tin Hong Kong
| | - An Hong
- Department of Cell Biology Jinan University Guangzhou China
| | - Yihai Cao
- Department of Microbiology Tumor and Cell Biology Karolinska Institute Stockholm Sweden
| | - Wencai Ye
- College of Pharmacy Jinan University Guangzhou China
| | - Dongmei Zhang
- College of Pharmacy Jinan University Guangzhou China
| |
Collapse
|
32
|
Liu L, O’Kelly D, Schuetze R, Carlson G, Zhou H, Trawick ML, Pinney KG, Mason RP. Non-Invasive Evaluation of Acute Effects of Tubulin Binding Agents: A Review of Imaging Vascular Disruption in Tumors. Molecules 2021; 26:2551. [PMID: 33925707 PMCID: PMC8125421 DOI: 10.3390/molecules26092551] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022] Open
Abstract
Tumor vasculature proliferates rapidly, generally lacks pericyte coverage, and is uniquely fragile making it an attractive therapeutic target. A subset of small-molecule tubulin binding agents cause disaggregation of the endothelial cytoskeleton leading to enhanced vascular permeability generating increased interstitial pressure. The resulting vascular collapse and ischemia cause downstream hypoxia, ultimately leading to cell death and necrosis. Thus, local damage generates massive amplification and tumor destruction. The tumor vasculature is readily accessed and potentially a common target irrespective of disease site in the body. Development of a therapeutic approach and particularly next generation agents benefits from effective non-invasive assays. Imaging technologies offer varying degrees of sophistication and ease of implementation. This review considers technological strengths and weaknesses with examples from our own laboratory. Methods reveal vascular extent and patency, as well as insights into tissue viability, proliferation and necrosis. Spatiotemporal resolution ranges from cellular microscopy to single slice tomography and full three-dimensional views of whole tumors and measurements can be sufficiently rapid to reveal acute changes or long-term outcomes. Since imaging is non-invasive, each tumor may serve as its own control making investigations particularly efficient and rigorous. The concept of tumor vascular disruption was proposed over 30 years ago and it remains an active area of research.
Collapse
Affiliation(s)
- Li Liu
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.L.); (D.O.); (R.S.); (H.Z.)
| | - Devin O’Kelly
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.L.); (D.O.); (R.S.); (H.Z.)
| | - Regan Schuetze
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.L.); (D.O.); (R.S.); (H.Z.)
| | - Graham Carlson
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (G.C.); (M.L.T.); (K.G.P.)
| | - Heling Zhou
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.L.); (D.O.); (R.S.); (H.Z.)
| | - Mary Lynn Trawick
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (G.C.); (M.L.T.); (K.G.P.)
| | - Kevin G. Pinney
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (G.C.); (M.L.T.); (K.G.P.)
| | - Ralph P. Mason
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.L.); (D.O.); (R.S.); (H.Z.)
| |
Collapse
|
33
|
Bao X, Shen N, Lou Y, Yu H, Wang Y, Liu L, Tang Z, Chen X. Enhanced anti-PD-1 therapy in hepatocellular carcinoma by tumor vascular disruption and normalization dependent on combretastatin A4 nanoparticles and DC101. Theranostics 2021; 11:5955-5969. [PMID: 33897892 PMCID: PMC8058708 DOI: 10.7150/thno.58164] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/23/2021] [Indexed: 12/23/2022] Open
Abstract
Anti-programmed cell death protein 1 (PD-1) therapy has shown promising efficacy in hepatocellular carcinoma (HCC), but its response rates in advanced HCC are lower than 20%. A critical reason for this is the imbalance between CD8+ T cells and tumor burden. Here, a novel concept of vascular disruption and normalization dependent on a polymeric vascular disrupting agent (VDA) poly (L-glutamic acid)-graft-methoxy poly (ethylene glycol)/combretastatin A4 (CA4-NPs) + a vascular endothelial growth factor (VEGF)/VEGF receptor 2 (VEGFR2) inhibitor DC101 is applied to improve anti-PD-1 therapy, wherein CA4-NPs reduce tumor burden and DC101 simultaneously increases the number of intratumoral CD8+ T cells, successfully regulating the abovementioned imbalance in an H22 tumor model. Methods: Blood vessel density, tumor cell proliferation, and necrosis were evaluated to reveal the effects on reducing tumor burden by CA4-NP treatment. Pericyte coverage of blood vessels, tumor blood vessel perfusion, tumor hypoxia, and intratumoral immune cells were examined to verify their role in vascular normalization and immune cell homing of DC101. Furthermore, the effects of CA4-NPs + DC101 on reducing tumor burden and increasing the number of immune cells were studied. Finally, tumor suppression, intratumoral CD8+ T cell activation, and the synergistic effects of anti-PD-1 combined with CA4-NPs + DC101 were verified. Results: The tumor inhibition rate of anti-PD-1 antibody combined with CA4-NPs + DC101 reached 86.4%, which was significantly higher than that of anti-PD-1 (16.8%) alone. Importantly, the Q value reflecting the synergy between CA4-NPs + DC101 and anti-PD-1 was 1.24, demonstrating a strong synergistic effect. Furthermore, CA4-NPs + DC101 improved anti-PD-1 therapy by increasing the number of intratumoral CD8+ T cells (anti-PD-1, 0.31% vs triple drug combination, 1.18%). Conclusion: These results reveal a novel approach to enhance anti-PD-1 therapy with VDAs + VEGF/VEGFR2 inhibitors in HCC.
Collapse
Affiliation(s)
- Xin Bao
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun 130041, P. R. China
- Department of Thyroid, The Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Na Shen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Yan Lou
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun, 130041, P. R. China
| | - Haiyang Yu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Yue Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Linlin Liu
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| |
Collapse
|
34
|
Hoque MM, Abdelazim H, Jenkins-Houk C, Wright D, Patel BM, Chappell JC. The cerebral microvasculature: Basic and clinical perspectives on stroke and glioma. Microcirculation 2021; 28:e12671. [PMID: 33171539 PMCID: PMC11064683 DOI: 10.1111/micc.12671] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/13/2020] [Accepted: 11/04/2020] [Indexed: 12/12/2022]
Abstract
Microvascular networks are vital components of the cardiovascular system, performing many key roles in maintaining the health and homeostasis of the tissues and organs in which they develop. As discussed in this review, the molecular and cellular components within the microcirculation orchestrate critical processes to establish functional capillary beds, including organization of endothelial cell (EC) polarity, guiding investment of vascular pericytes (PCs), and building the specialized extracellular matrix (ECM) that comprises the vascular basement membrane (vBM). Herein, we further discuss the unique features of the microvasculature in the central nervous system (CNS), focusing on the cells contributing to the neurovascular unit (NVU) that form and maintain the blood-brain barrier (BBB). With a focus on vascular PCs, we offer basic and clinical perspectives on neurovascular-related pathologies that involve defects within the cerebral microvasculature. Specifically, we present microvascular anomalies associated with glioblastoma multiforme (GBM) including defects in vascular-immune cell interactions and associated clinical therapies targeting microvessels (ie, vascular-disrupting/anti-angiogenic agents and focused ultrasound). We also discuss the involvement of the microcirculation in stroke responses and potential therapeutic approaches. Our goal was to compare the cellular and molecular changes that occur in the microvasculature and NVU, and to provide a commentary on factors driving disease progression in GBM and stroke. We conclude with a forward-looking perspective on the importance of microcirculation research in developing clinical treatments for these devastating conditions.
Collapse
Affiliation(s)
- Maruf M. Hoque
- Center for Heart and Reparative Medicine Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
- Graduate Program in Translational Biology, Medicine and Health, Virginia Tech, Blacksburg, VA 24061, USA
| | - Hanaa Abdelazim
- Center for Heart and Reparative Medicine Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
- Graduate Program in Translational Biology, Medicine and Health, Virginia Tech, Blacksburg, VA 24061, USA
| | | | - Dawn Wright
- Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA
| | - Biraj M. Patel
- Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA
- Department of Radiology, Carilion Clinic, Roanoke, VA, 24016, USA
| | - John C. Chappell
- Center for Heart and Reparative Medicine Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
35
|
Abstract
Fibroblast activation protein-α (FAP) is a type-II transmembrane serine protease expressed almost exclusively to pathological conditions including fibrosis, arthritis, and cancer. Across most cancer types, elevated FAP is associated with worse clinical outcomes. Despite the clear association between FAP and disease severity, the biological reasons underlying these clinical observations remain unclear. Here we review basic FAP biology and FAP's role in non-oncologic and oncologic disease. We further explore how FAP may worsen clinical outcomes via its effects on extracellular matrix remodeling, intracellular signaling regulation, angiogenesis, epithelial-to-mesenchymal transition, and immunosuppression. Lastly, we discuss the potential to exploit FAP biology to improve clinical outcomes.
Collapse
Affiliation(s)
- Allison A Fitzgerald
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3870 Reservoir Road NW, Washington, DC, 20057, USA
| | - Louis M Weiner
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3870 Reservoir Road NW, Washington, DC, 20057, USA.
| |
Collapse
|
36
|
Li X, Chen M, Lu W, Tang J, Deng L, Wen Q, Huang M, Deng R, Ye G, Ye W, Zhang D. Targeting FAPα-expressing tumor-associated mesenchymal stromal cells inhibits triple-negative breast cancer pulmonary metastasis. Cancer Lett 2021; 503:32-42. [PMID: 33482262 DOI: 10.1016/j.canlet.2021.01.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/08/2020] [Accepted: 01/13/2021] [Indexed: 02/08/2023]
Abstract
Tumor metastasis is the main cause of death in patients with triple-negative breast cancer (TNBC). Bone marrow-derived mesenchymal stem cells (BM-MSCs) have tropism towards tumor tissues, and can be converted into tumor-associated mesenchymal stromal cells (TA-MSCs) to facilitate TNBC metastasis through interactions with tumor-associated macrophages (TAMs). However, the underlying molecular mechanisms are complex and unclear, and effective strategies to suppress tumor metastasis via eliminating TA-MSCs are still lacking. Here, we demonstrate that fibroblast activation protein alpha (FAPα) was overexpressed in TA-MSCs, which prompts TA-MSCs to secrete multiple C-C motif chemokine ligands, promoting C-C motif chemokine receptor 2 (CCR2)+ TAM recruitment and facilitating TAM polarization into the M2 phenotype, thereby promoting TNBC pulmonary metastasis. Z-GP-DAVLBH, an FAPα-activated vinblastine prodrug, induces FAPα+ TA-MSC apoptosis, which significantly suppresses CCR2+ TAM recruitment and polarization, thus inhibiting pulmonary metastasis of orthotopic TNBC cell-derived xenografts and patient-derived xenografts. This study provides insight into an important role of FAPα in mediating TA-MSC-induced TNBC metastasis and provides compelling evidence that targeting TA-MSCs with an FAPα-activated prodrug is a promising strategy for suppressing TNBC metastasis.
Collapse
Affiliation(s)
- Xiaobo Li
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Minfeng Chen
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Weijin Lu
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Jun Tang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China; Department of Breast Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Lijuan Deng
- Formula-pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, PR China
| | - Qing Wen
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Maohua Huang
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Rong Deng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060, PR China
| | - Geni Ye
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Wencai Ye
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China.
| | - Dongmei Zhang
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China.
| |
Collapse
|
37
|
Abstract
Heparanase is the only mammalian enzyme that cleaves heparan sulphate, an important component of the extracellular matrix. This leads to the remodelling of the extracellular matrix, whilst liberating growth factors and cytokines bound to heparan sulphate. This in turn promotes both physiological and pathological processes such as angiogenesis, immune cell migration, inflammation, wound healing and metastasis. Furthermore, heparanase exhibits non-enzymatic actions in cell signalling and in regulating gene expression. Cancer is underpinned by key characteristic features that promote malignant growth and disease progression, collectively termed the 'hallmarks of cancer'. Essentially, all cancers examined to date have been reported to overexpress heparanase, leading to enhanced tumour growth and metastasis with concomitant poor patient survival. With its multiple roles within the tumour microenvironment, heparanase has been demonstrated to regulate each of these hallmark features, in turn highlighting the need for heparanase-targeted therapies. However, recent discoveries which demonstrated that heparanase can also regulate vital anti-tumour mechanisms have cast doubt on this approach. This review will explore the myriad ways by which heparanase functions as a key regulator of the hallmarks of cancer and will highlight its role as a major component within the tumour microenvironment. The dual role of heparanase within the tumour microenvironment, however, emphasises the need for further investigation into defining its precise mechanism of action in different cancer settings.
Collapse
Affiliation(s)
- Krishnath M Jayatilleke
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Plenty Road & Kingsbury Drive, Melbourne, VIC, 3086, Australia
| | - Mark D Hulett
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Plenty Road & Kingsbury Drive, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
38
|
Brennen WN, J Thorek DL, Jiang W, Krueger TE, Antony L, Denmeade SR, Isaacs JT. Overcoming stromal barriers to immuno-oncological responses via fibroblast activation protein-targeted therapy. Immunotherapy 2020; 13:155-175. [PMID: 33148078 DOI: 10.2217/imt-2020-0066] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The tumor microenvironment contributes to disease progression through multiple mechanisms, including immune suppression mediated in part by fibroblast activation protein (FAP)-expressing cells. Herein, a review of FAP biology is presented, supplemented with primary data. This includes FAP expression in prostate cancer and activation of latent reservoirs of TGF-β and VEGF to produce a positive feedback loop. This collectively suggests a normal wound repair process subverted during cancer pathophysiology. There has been immense interest in targeting FAP for diagnostic, monitoring and therapeutic purposes. Until recently, this development has outpaced an understanding of the biology; impeding optimal translation into the clinic. A summary of these applications is provided with an emphasis on eliminating tumor-infiltrating FAP-positive cells to overcome stromal barriers to immuno-oncological responses.
Collapse
Affiliation(s)
- W Nathaniel Brennen
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, MD 21287, USA
| | - Daniel L J Thorek
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO 63310, USA.,Department of Biomedical Engineering, Washington University School of Medicine, Saint Louis, MO 63310, USA
| | - Wen Jiang
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Timothy E Krueger
- Department of Pharmacology & Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Lizamma Antony
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, MD 21287, USA
| | - Samuel R Denmeade
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, MD 21287, USA
| | - John T Isaacs
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, MD 21287, USA
| |
Collapse
|
39
|
Li J, Burgess DJ. Nanomedicine-based drug delivery towards tumor biological and immunological microenvironment. Acta Pharm Sin B 2020; 10:2110-2124. [PMID: 33304781 PMCID: PMC7714990 DOI: 10.1016/j.apsb.2020.05.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/15/2020] [Accepted: 05/21/2020] [Indexed: 12/17/2022] Open
Abstract
The complex tumor microenvironment is a most important factor in cancer development. The biological microenvironment is composed of a variety of barriers including the extracellular matrix and associated cells such as endothelia cells, pericytes, and cancer-associated fibroblasts. Different strategies can be utilized to enhance nanoparticle-based drug delivery and distribution into tumor tissues addressing the extracellular matrix or cellular components. In addition to the biological microenvironment, the immunological conditions around the tumor tissue can be very complicated and cancer cells have various ways of evading immune surveillance. Nanoparticle drug delivery systems can enhance cancer immunotherapy by tuning the immunological response and memory of various immune cells such as T cells, B cells, macrophages, and dendritic cells. In this review, the main components in the tumor biological and immunological environment are discussed. The focus is on recent advances in nanoparticle-based drug delivery systems towards targets within the tumor microenvironment to improve cancer chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Jin Li
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269-3092, USA
| | - Diane J. Burgess
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269-3092, USA
| |
Collapse
|
40
|
Smolarczyk R, Czapla J, Jarosz-Biej M, Czerwinski K, Cichoń T. Vascular disrupting agents in cancer therapy. Eur J Pharmacol 2020; 891:173692. [PMID: 33130277 DOI: 10.1016/j.ejphar.2020.173692] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 10/15/2020] [Accepted: 10/26/2020] [Indexed: 12/14/2022]
Abstract
Tumor blood vessel formation is a key process for tumor expansion. Tumor vessels are abnormal and differ from normal vessels in architecture and components. Besides oxygen and nutrients supply, the tumor vessels system, due to its abnormality, is responsible for: hypoxia formation, and metastatic routes. Tumor blood vessels can be a target of anti-cancer therapies. There are two types of therapies that target tumor vessels. The first one is the inhibition of the angiogenesis process. However, the inhibition is often ineffective because of alternative angiogenesis mechanism activation. The second type is a specific targeting of existing tumor blood vessels by vascular disruptive agents (VDAs). There are three groups of VDAs: microtubule destabilizing drugs, flavonoids with anti-vascular functions, and tumor vascular targeted drugs based on endothelial cell receptors. However, VDAs possess some limitations. They may be cardiotoxic and their application in therapy may leave viable residual, so called, rim cells on the edge of the tumor. However, it seems that a well-designed combination of VDAs with other anti-cancer drugs may bring a significant therapeutic effect. In this article, we describe three groups of vascular disruptive agents with their advantages and disadvantages. We mention VDAs clinical trials. Finally, we present the current possibilities of VDAs combination with other anti-cancer drugs.
Collapse
Affiliation(s)
- Ryszard Smolarczyk
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland.
| | - Justyna Czapla
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland.
| | - Magdalena Jarosz-Biej
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland.
| | - Kyle Czerwinski
- University of Manitoba, Faculty of Science. 66 Chancellors Cir, Winnipeg, Canada.
| | - Tomasz Cichoń
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej Street 15, 44-102, Gliwice, Poland.
| |
Collapse
|
41
|
Ebert LM, Yu W, Gargett T, Toubia J, Kollis PM, Tea MN, Ebert BW, Bardy C, van den Hurk M, Bonder CS, Manavis J, Ensbey KS, Oksdath Mansilla M, Scheer KG, Perrin SL, Ormsby RJ, Poonnoose S, Koszyca B, Pitson SM, Day BW, Gomez GA, Brown MP. Endothelial, pericyte and tumor cell expression in glioblastoma identifies fibroblast activation protein (FAP) as an excellent target for immunotherapy. Clin Transl Immunology 2020; 9:e1191. [PMID: 33082953 PMCID: PMC7557106 DOI: 10.1002/cti2.1191] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/07/2020] [Accepted: 09/14/2020] [Indexed: 12/12/2022] Open
Abstract
Objectives Targeted immunotherapies such as chimeric antigen receptor (CAR)-T cells are emerging as attractive treatment options for glioblastoma, but rely on identification of a suitable tumor antigen. We validated a new target antigen for glioblastoma, fibroblast activation protein (FAP), by undertaking a detailed expression study of human samples. Methods Glioblastoma and normal tissues were assessed using immunostaining, supported by analyses of published transcriptomic datasets. Short-term cultures of glioma neural stem (GNS) cells were compared to cultures of healthy astrocytes and neurons using flow cytometry. Glioblastoma tissues were dissociated and analysed by high-parameter flow cytometry and single-cell transcriptomics (scRNAseq). Results Compared to normal brain, FAP was overexpressed at the gene and protein level in a large percentage of glioblastoma tissues, with highest levels of expression associated with poorer prognosis. FAP was also overexpressed in several paediatric brain cancers. FAP was commonly expressed by cultured GNS cells but absent from normal neurons and astrocytes. Within glioblastoma tissues, the strongest expression of FAP was around blood vessels. In fact, almost every tumor vessel was highlighted by FAP expression, whereas normal tissue vessels and cultured endothelial cells (ECs) lacked expression. Single-cell analyses of dissociated tumors facilitated a detailed characterisation of the main cellular components of the glioblastoma microenvironment and revealed that vessel-localised FAP is because of expression on both ECs and pericytes. Conclusion Fibroblast activation protein is expressed by multiple cell types within glioblastoma, highlighting it as an ideal immunotherapy antigen to target destruction of both tumor cells and their supporting vascular network.
Collapse
Affiliation(s)
- Lisa M Ebert
- Centre for Cancer Biology SA Pathology and University of South Australia Adelaide Australia.,Adelaide Medical School University of Adelaide Adelaide Australia
| | - Wenbo Yu
- Centre for Cancer Biology SA Pathology and University of South Australia Adelaide Australia
| | - Tessa Gargett
- Centre for Cancer Biology SA Pathology and University of South Australia Adelaide Australia.,Adelaide Medical School University of Adelaide Adelaide Australia
| | - John Toubia
- Centre for Cancer Biology SA Pathology and University of South Australia Adelaide Australia
| | - Paris M Kollis
- Centre for Cancer Biology SA Pathology and University of South Australia Adelaide Australia.,Adelaide Medical School University of Adelaide Adelaide Australia
| | - Melinda N Tea
- Centre for Cancer Biology SA Pathology and University of South Australia Adelaide Australia
| | - Brenton W Ebert
- Centre for Cancer Biology SA Pathology and University of South Australia Adelaide Australia
| | - Cedric Bardy
- South Australian Health and Medical Research Institute (SAHMRI) Adelaide Australia.,College of Medicine & Public Health Flinders University Adelaide Australia
| | - Mark van den Hurk
- South Australian Health and Medical Research Institute (SAHMRI) Adelaide Australia.,College of Medicine & Public Health Flinders University Adelaide Australia
| | - Claudine S Bonder
- Centre for Cancer Biology SA Pathology and University of South Australia Adelaide Australia.,Adelaide Medical School University of Adelaide Adelaide Australia
| | - Jim Manavis
- Adelaide Medical School University of Adelaide Adelaide Australia
| | - Kathleen S Ensbey
- Department of Cell and Molecular Biology Sid Faithfull Brain Cancer Laboratory QIMR Berghofer Medical Research Institute Brisbane QLD Australia
| | | | - Kaitlin G Scheer
- Centre for Cancer Biology SA Pathology and University of South Australia Adelaide Australia.,Clinical and Health Sciences University of South Australia Adelaide Australia
| | - Sally L Perrin
- Centre for Cancer Biology SA Pathology and University of South Australia Adelaide Australia.,Clinical and Health Sciences University of South Australia Adelaide Australia
| | - Rebecca J Ormsby
- College of Medicine & Public Health Flinders University Adelaide Australia
| | - Santosh Poonnoose
- College of Medicine & Public Health Flinders University Adelaide Australia.,Department of Neurosurgery Flinders Medical Centre Bedford Park Australia
| | - Barbara Koszyca
- Department of Anatomical Pathology SA Pathology Adelaide Australia
| | - Stuart M Pitson
- Centre for Cancer Biology SA Pathology and University of South Australia Adelaide Australia.,Adelaide Medical School University of Adelaide Adelaide Australia
| | - Bryan W Day
- Department of Cell and Molecular Biology Sid Faithfull Brain Cancer Laboratory QIMR Berghofer Medical Research Institute Brisbane QLD Australia.,Faculty of Health Queensland University of Technology Brisbane QLD Australia.,Faculty of Medicine The University of Queensland Brisbane QLD Australia
| | - Guillermo A Gomez
- Centre for Cancer Biology SA Pathology and University of South Australia Adelaide Australia
| | - Michael P Brown
- Centre for Cancer Biology SA Pathology and University of South Australia Adelaide Australia.,Adelaide Medical School University of Adelaide Adelaide Australia.,Cancer Clinical Trials Unit Royal Adelaide Hospital Adelaide Australia
| |
Collapse
|
42
|
Yetkin-Arik B, Kastelein AW, Klaassen I, Jansen CHJR, Latul YP, Vittori M, Biri A, Kahraman K, Griffioen AW, Amant F, Lok CAR, Schlingemann RO, van Noorden CJF. Angiogenesis in gynecological cancers and the options for anti-angiogenesis therapy. Biochim Biophys Acta Rev Cancer 2020; 1875:188446. [PMID: 33058997 DOI: 10.1016/j.bbcan.2020.188446] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 10/02/2020] [Accepted: 10/04/2020] [Indexed: 02/06/2023]
Abstract
Angiogenesis is required in cancer, including gynecological cancers, for the growth of primary tumors and secondary metastases. Development of anti-angiogenesis therapy in gynecological cancers and improvement of its efficacy have been a major focus of fundamental and clinical research. However, survival benefits of current anti-angiogenic agents, such as bevacizumab, in patients with gynecological cancer, are modest. Therefore, a better understanding of angiogenesis and the tumor microenvironment in gynecological cancers is urgently needed to develop more effective anti-angiogenic therapies, either or not in combination with other therapeutic approaches. We describe the molecular aspects of (tumor) blood vessel formation and the tumor microenvironment and provide an extensive clinical overview of current anti-angiogenic therapies for gynecological cancers. We discuss the different phenotypes of angiogenic endothelial cells as potential therapeutic targets, strategies aimed at intervention in their metabolism, and approaches targeting their (inflammatory) tumor microenvironment.
Collapse
Affiliation(s)
- Bahar Yetkin-Arik
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands; Department of Medical Biology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Arnoud W Kastelein
- Department of Obstetrics and Gynaecology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands.
| | - Ingeborg Klaassen
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands; Department of Medical Biology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Charlotte H J R Jansen
- Department of Obstetrics and Gynaecology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Yani P Latul
- Department of Obstetrics and Gynaecology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands
| | - Miloš Vittori
- Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Aydan Biri
- Department of Obstetrics and Gynecology, Koru Ankara Hospital, Ankara, Turkey
| | - Korhan Kahraman
- Department of Obstetrics and Gynecology, Bahcesehir University School of Medicine, Istanbul, Turkey
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Frederic Amant
- Department of Oncology, KU Leuven, Leuven, Belgium; Center for Gynaecological Oncology, Antoni van Leeuwenhoek, Amsterdam, the Netherlands; Center for Gynaecological Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Center for Gynaecological Oncology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Christianne A R Lok
- Center for Gynaecological Oncology, Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Reinier O Schlingemann
- Ocular Angiogenesis Group, Department of Ophthalmology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands; Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Cornelis J F van Noorden
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, the Netherlands; Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| |
Collapse
|
43
|
Yuan W, Hai B, Ren X, Zhu J, Zhang C, Guan Z, Jia J, Wang H, Cao B, Song C. Single-dose local intraosseous injection of simvastatin suppresses breast cancer with tumor vascular normalization. Transl Oncol 2020; 13:100867. [PMID: 32950929 PMCID: PMC7509234 DOI: 10.1016/j.tranon.2020.100867] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 08/05/2020] [Accepted: 08/07/2020] [Indexed: 12/23/2022] Open
Abstract
Tumor vessels play important roles in cancer development and angiogenesis has been characterized as an essential process for tumor cell tumor growth. Our previous studies found that a single-dose local intraosseous simvastatin injection rapidly and long-termly mobilized bone marrow-derived endothelial progenitor cells to peripheral blood, promoting angiogenesis and ameliorating ischemia injury. However, whether intraosseous injection of simvastatin participates in cancer progression and the role of angiogenesis enhancement in this process remain unknown. In this study, we found that intraosseous injection of simvastatin improves tumor vascular structure, along with increasing the percentage of pericyte coverage on tumor vessels, and reducing vascular permeability, tumor hypoxia and tumor necrosis. Further, we demonstrate that a single-dose local intraosseous simvastatin injection suppresses tumor growth, facilitates sensitivity of chemotherapy and prolongs survival in breast cancer-bearing mice. In addition, oral application, intravenous, subcutaneous and intraperitoneal injection of simvastatin do not show these effects. Taken together, these results demonstrate that intraosseous injection of simvastatin suppresses breast cancer with tumor vascular normalization, which might be a promising strategy for cancer treatment.
Collapse
Affiliation(s)
- Wanqiong Yuan
- Department of Orthopedics, Peking University Third Hospital, Beijing Key Laboratory of Spinal Disease, Beijing, China.
| | - Bao Hai
- Department of Orthopedics, Peking University Third Hospital, Beijing Key Laboratory of Spinal Disease, Beijing, China.
| | - Xiaoqing Ren
- Department of Pharmacy, Peking University Third Hospital, Beijing, China
| | - Junxiong Zhu
- Department of Orthopedics, Peking University Third Hospital, Beijing Key Laboratory of Spinal Disease, Beijing, China.
| | - Chenggui Zhang
- Department of Orthopedics, Peking University Third Hospital, Beijing Key Laboratory of Spinal Disease, Beijing, China.
| | - Zhiyuan Guan
- Department of Orthopedics, Peking University Third Hospital, Beijing Key Laboratory of Spinal Disease, Beijing, China.
| | - Jialin Jia
- Department of Orthopedics, Peking University Third Hospital, Beijing Key Laboratory of Spinal Disease, Beijing, China.
| | - Hong Wang
- Department of Orthopedics, Peking University Third Hospital, Beijing Key Laboratory of Spinal Disease, Beijing, China
| | - Baoshan Cao
- Department of Tumor Chemotherapy and Radiation Sickness, Peking University Third Hospital, Beijing, China
| | - Chunli Song
- Department of Orthopedics, Peking University Third Hospital, Beijing Key Laboratory of Spinal Disease, Beijing, China.
| |
Collapse
|
44
|
Lei X, Zhong Y, Huang L, Li S, Fu J, Zhang L, Zhang Y, Deng Q, Yu X. Identification of a novel tumor angiogenesis inhibitor targeting Shh/Gli1 signaling pathway in Non-small cell lung cancer. Cell Death Dis 2020; 11:232. [PMID: 32286274 PMCID: PMC7156472 DOI: 10.1038/s41419-020-2425-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 02/02/2023]
Abstract
Although angiogenesis inhibitors targeting VEGF/VEGFR2 have been applied for tumor therapy, the outcomes are still unsatisfactory. Thus, it is urgent to develop novel angiogenesis inhibitor for cancer therapy from new perspectives. Identification of novel angiogenesis inhibitor from natural products is believed to be one of most promising strategy. In this study, we showed that pristimerin, an active agent isolated from traditional Chinese herbal medicine Celastrus aculeatus Merr, was a novel tumor angiogenesis inhibitor that targeting sonic hedgehog (Shh)/glioma associated oncogene 1 (Gli1) signaling pathway in non-small cell lung cancer (NSCLC). We showed that pristimerin affected both the early- and late-stage of angiogenesis, suggesting by that pristimerin inhibited Shh-induced endothelial cells proliferation, migration, invasion as well as pericytes recruitment to the endothelial tubes, which is critical for the new blood vessel maturation. It also suppressed tube formation, vessel sprouts formation and neovascularization in chicken embryo chorioallantoic membrane (CAM). Moreover, it significantly decreased microvessel density (MVD) and pericyte coverage in NCI-H1299 xenografts, resulting in tumor growth inhibition. Further research revealed that pristimerin suppressed tumor angiogenesis by inhibiting the nucleus distribution of Gli1, leading to inactivation of Shh/Gli1 and its downstream signaling pathway. Taken together, our study showed that pristimerin was a promising novel anti-angiogenic agent for the NSCLC therapy and targeting Shh/Gli1 signaling pathway was an effective approach to suppress tumor angiogenesis.
Collapse
Affiliation(s)
- Xueping Lei
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, Guangdong, China
| | - Yihang Zhong
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, Guangdong, China
| | - Lijuan Huang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, Guangdong, China
| | - Songpei Li
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, Guangdong, China
| | - Jijun Fu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, Guangdong, China
| | - Lingmin Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, Guangdong, China
| | - Yu Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, Guangdong, China
| | - Qiudi Deng
- GMU-GIBH Joint School of Life Sciences & the Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Xiyong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, Guangdong, China.
| |
Collapse
|
45
|
Yu H, Shen N, Bao Y, Chen L, Tang Z. Tumor regression and potentiation of polymeric vascular disrupting therapy through reprogramming of a hypoxia microenvironment with temsirolimus. Biomater Sci 2020; 8:325-332. [DOI: 10.1039/c9bm01398a] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
To relieve a tumor hypoxia microenvironment, the mTOR inhibitor temsirolimus was employed to modulate the tumor microenvironment when treated with CA4-NPs.
Collapse
Affiliation(s)
- Haiyang Yu
- Department of Chemistry
- Northeast Normal University
- Changchun 130024
- People's Republic of China
- Key Laboratory of Polymer Ecomaterials
| | - Na Shen
- Key Laboratory of Polymer Ecomaterials
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- People's Republic of China
| | - Yanli Bao
- Department of Chemistry
- Northeast Normal University
- Changchun 130024
- People's Republic of China
| | - Li Chen
- Department of Chemistry
- Northeast Normal University
- Changchun 130024
- People's Republic of China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- People's Republic of China
| |
Collapse
|
46
|
Caporarello N, D’Angeli F, Cambria MT, Candido S, Giallongo C, Salmeri M, Lombardo C, Longo A, Giurdanella G, Anfuso CD, Lupo G. Pericytes in Microvessels: From "Mural" Function to Brain and Retina Regeneration. Int J Mol Sci 2019; 20:ijms20246351. [PMID: 31861092 PMCID: PMC6940987 DOI: 10.3390/ijms20246351] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/13/2019] [Accepted: 12/14/2019] [Indexed: 12/13/2022] Open
Abstract
Pericytes are branched cells located in the wall of capillary blood vessels that are found throughout the body, embedded within the microvascular basement membrane and wrapping endothelial cells, with which they establish a strong physical contact. Pericytes regulate angiogenesis, vessel stabilization, and contribute to the formation of both the blood-brain and blood-retina barriers by Angiopoietin-1/Tie-2, platelet derived growth factor (PDGF) and transforming growth factor (TGF) signaling pathways, regulating pericyte-endothelial cell communication. Human pericytes that have been cultured for a long period give rise to multilineage progenitor cells and exhibit mesenchymal stem cell (MSC) features. We focused our attention on the roles of pericytes in brain and ocular diseases. In particular, pericyte involvement in brain ischemia, brain tumors, diabetic retinopathy, and uveal melanoma is described. Several molecules, such as adenosine and nitric oxide, are responsible for pericyte shrinkage during ischemia-reperfusion. Anti-inflammatory molecules, such as IL-10, TGFβ, and MHC-II, which are increased in glioblastoma-activated pericytes, are responsible for tumor growth. As regards the eye, pericytes play a role not only in ocular vessel stabilization, but also as a stem cell niche that contributes to regenerative processes in diabetic retinopathy. Moreover, pericytes participate in melanoma cell extravasation and the genetic ablation of the PDGF receptor reduces the number of pericytes and aberrant tumor microvessel formation with important implications for therapy efficacy. Thanks to their MSC features, pericytes could be considered excellent candidates to promote nervous tissue repair and for regenerative medicine.
Collapse
Affiliation(s)
- Nunzia Caporarello
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA;
| | - Floriana D’Angeli
- Section of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (F.D.); (M.T.C.); (A.L.); (G.G.)
| | - Maria Teresa Cambria
- Section of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (F.D.); (M.T.C.); (A.L.); (G.G.)
| | - Saverio Candido
- Section of General and Clinical Pathology and Oncology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy;
| | - Cesarina Giallongo
- Section of Haematology, Department of General Surgery and Medical-Surgical Specialties, University of Catania, 95123 Catania, Italy;
| | - Mario Salmeri
- Section of Microbiology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (M.S.); (C.L.)
| | - Cinzia Lombardo
- Section of Microbiology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (M.S.); (C.L.)
| | - Anna Longo
- Section of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (F.D.); (M.T.C.); (A.L.); (G.G.)
| | - Giovanni Giurdanella
- Section of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (F.D.); (M.T.C.); (A.L.); (G.G.)
| | - Carmelina Daniela Anfuso
- Section of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (F.D.); (M.T.C.); (A.L.); (G.G.)
- Correspondence: (G.L.); (C.D.A.); Tel.: +39-095-4781158 (G.L.); +39-095-4781170 (C.D.A.)
| | - Gabriella Lupo
- Section of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (F.D.); (M.T.C.); (A.L.); (G.G.)
- Correspondence: (G.L.); (C.D.A.); Tel.: +39-095-4781158 (G.L.); +39-095-4781170 (C.D.A.)
| |
Collapse
|
47
|
Johnson SP, Ogunlade O, Lythgoe MF, Beard P, Pedley RB. Longitudinal Photoacoustic Imaging of the Pharmacodynamic Effect of Vascular Targeted Therapy on Tumors. Clin Cancer Res 2019; 25:7436-7447. [PMID: 31551349 PMCID: PMC7611302 DOI: 10.1158/1078-0432.ccr-19-0360] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 07/29/2019] [Accepted: 09/19/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Photoacoustic imaging (PAI) is a novel noninvasive and nonionizing imaging technique that allows longitudinal imaging of tumor vasculature in vivo and monitoring of response to therapy, especially for vascular targeted chemotherapy agents. In this study, we used a novel high-resolution all-optical PAI scanner to observe the pharmacodynamic response to the vascular-disrupting agent OXi4503. EXPERIMENTAL DESIGN Two models of colorectal carcinoma (SW1222 and LS174T) that possess differing pathophysiologic vascularization were established as subcutaneous tumors in mice. Monitoring of response was performed over a 16-day "regrowth" period following treatment at 40 mg/kg, and at day 2 for a "dose response" study at 40 mg/kg, 10 mg/kg, 1 mg/kg, and sham dose. RESULTS Qualitative and quantitative changes in PA signal are observed, with an initial decrease followed by a plateau and subsequent return of signal indicating regrowth. Both tumor types exhibited a decrease in signal; however, the more vascularized SW1222 tumors show greater response to treatment. Decreasing the dose of OXi4503 led to a decrease in PA signal intensity of 60%, 52%, and 20% in SW1222 tumors and 30%, 26%, and 4% for LS174T tumors. CONCLUSIONS We have shown for the first time that PAI can observe the pharmacodynamic response of tumor vasculature to drug treatment both longitudinally and at different dose levels. Assessment of differing response to treatment based on vascular pathophysiologic differences among patients has the potential to provide personalized drug therapy; we have demonstrated that PAI, which is clinically translatable, could be a powerful tool for this purpose.
Collapse
Affiliation(s)
- S Peter Johnson
- UCL Cancer Institute, University College London, London, United Kingdom.
| | - Olumide Ogunlade
- UCL Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Mark F Lythgoe
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, United Kingdom
| | - Paul Beard
- UCL Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - R Barbara Pedley
- UCL Cancer Institute, University College London, London, United Kingdom
| |
Collapse
|
48
|
Delahousse J, Skarbek C, Paci A. Prodrugs as drug delivery system in oncology. Cancer Chemother Pharmacol 2019; 84:937-958. [DOI: 10.1007/s00280-019-03906-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 07/05/2019] [Indexed: 02/07/2023]
|
49
|
Lindner T, Loktev A, Giesel F, Kratochwil C, Altmann A, Haberkorn U. Targeting of activated fibroblasts for imaging and therapy. EJNMMI Radiopharm Chem 2019; 4:16. [PMID: 31659499 PMCID: PMC6658625 DOI: 10.1186/s41181-019-0069-0] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 07/16/2019] [Indexed: 02/07/2023] Open
Abstract
Tumors form a complex environment consisting of a variety of non-malignant cells. Especially cancer-associated fibroblasts have been shown to have an important role for different aspects of malignant tumors such as migration, metastasis, resistance to chemotherapy and immunosuppression. Therefore, a targeting of these cells may be useful for both imaging and therapy. In this respect, an interesting target is the fibroblast activation protein (FAP) which is expressed in activated fibroblasts, but not in quiescent fibroblasts, giving the opportunity to use this membrane-anchored enzyme as a target for radionuclide-based approaches for diagnosis and treatment of tumors and for the diagnosis of non-malignant disease associated with a remodelling of the extracellular matrix.
Collapse
Affiliation(s)
- Thomas Lindner
- Department of Nuclear Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Anastasia Loktev
- Department of Nuclear Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frederik Giesel
- Department of Nuclear Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Clemens Kratochwil
- Department of Nuclear Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Annette Altmann
- Department of Nuclear Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Uwe Haberkorn
- Department of Nuclear Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
50
|
Wang Y, Yu H, Zhang D, Wang G, Song W, Liu Y, Ma S, Tang Z, Liu Z, Sakurai K, Chen X. Co-administration of combretastatin A4 nanoparticles and sorafenib for systemic therapy of hepatocellular carcinoma. Acta Biomater 2019; 92:229-240. [PMID: 31100462 DOI: 10.1016/j.actbio.2019.05.028] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 05/07/2019] [Accepted: 05/09/2019] [Indexed: 12/26/2022]
Abstract
Effective systemic therapy is highly desired for the treatment of hepatocellular carcinoma (HCC). In this study, a combination of nanoparticles of poly(L-glutamic acid)-graft-methoxy poly(ethylene glycol)/combretastatin A4 sodium salt (CA4-NPs) plus sorafenib is developed for the cooperative systemic treatment of HCC. The CA4-NPs leads to the disruption of established tumor blood vessels and extensive tumor necrosis, however, inducing increased expression of VEGF-A and angiogenesis. Sorafenib reduces the VEGF-A induced angiogenesis and further inhibits tumor proliferation, cooperating with the CA4-NPs. A significant decrease in tumor volume and prolonged survival time are observed in the combination group of CA4-NPs plus sorafenib compared with CA4-NPs or sorafenib monotherapy in subcutaneous and orthotopic H22 hepatic tumor models. Seventy-one percent of the mice are alive without residual tumor at 96 days post tumor inoculation for the subcutaneous models treated with CA4-NPs 30 or 35 mg·kg-1 plus sorafenib 30 mg·kg-1. Our findings suggest that co-administration of sorafenib and CA4-NPs possesses significant antitumor efficacy for HCC treatment. STATEMENT OF SIGNIFICANCE: Effective systemic therapy is highly desired for the treatment of hepatocellular carcinoma (HCC). Herein, we demonstrate that a combination of nanoparticles of poly(L-glutamic acid)-graft-methoxy poly(ethylene glycol)/combretastatin A4 sodium salt (CA4-NPs) plus sorafenib is a promising synergistic approach for systemic treatment of HCC. The CA4-NPs leads to the disruption of established tumor blood vessels and extensive tumor necrosis, however, inducing increased expression of VEGF-A and angiogenesis. Sorafenib reduces the VEGF-A induced angiogenesis and further inhibits tumor proliferation, cooperating with the CA4-NPs.
Collapse
Affiliation(s)
- Yalin Wang
- Cancer Center, the First Hospital of Jilin University, Changchun 130021, China; Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Haiyang Yu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China
| | - Dawei Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Guanyi Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China
| | - Yingmin Liu
- Cancer Center, the First Hospital of Jilin University, Changchun 130021, China
| | - Sheng Ma
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China.
| | - Ziling Liu
- Cancer Center, the First Hospital of Jilin University, Changchun 130021, China.
| | - Kazuo Sakurai
- The University of Kitakyushu, Department of Chemistry and Biochemistry, 1-1, Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka 808-0135, Japan
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China
| |
Collapse
|