1
|
Li ZJ, He B, Domenichini A, Satiaputra J, Wood KH, Lakhiani DD, Bashaw AA, Nilsson LM, Li J, Bastow ER, Johansson-Percival A, Denisenko E, Forrest AR, Sakaram S, Carretero R, Hämmerling GJ, Nilsson JA, Lee GY, Ganss R. Pericyte phenotype switching alleviates immunosuppression and sensitizes vascularized tumors to immunotherapy in preclinical models. J Clin Invest 2024; 134:e179860. [PMID: 39286984 PMCID: PMC11405053 DOI: 10.1172/jci179860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/23/2024] [Indexed: 09/19/2024] Open
Abstract
T cell-based immunotherapies are a promising therapeutic approach for multiple malignancies, but their efficacy is limited by tumor hypoxia arising from dysfunctional blood vessels. Here, we report that cell-intrinsic properties of a single vascular component, namely the pericyte, contribute to the control of tumor oxygenation, macrophage polarization, vessel inflammation, and T cell infiltration. Switching pericyte phenotype from a synthetic to a differentiated state reverses immune suppression and sensitizes tumors to adoptive T cell therapy, leading to regression of melanoma in mice. In melanoma patients, improved survival is correlated with enhanced pericyte maturity. Importantly, pericyte plasticity is regulated by signaling pathways converging on Rho kinase activity, with pericyte maturity being inducible by selective low-dose therapeutics that suppress pericyte MEK, AKT, or notch signaling. We also show that low-dose targeted anticancer therapy can durably change the tumor microenvironment without inducing adaptive resistance, creating a highly translatable pathway for redosing anticancer targeted therapies in combination with immunotherapy to improve outcome.
Collapse
Affiliation(s)
- Zhi-Jie Li
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, Australia
- Department of Geriatric Medicine, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Bo He
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, Australia
| | - Alice Domenichini
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, Australia
| | - Jiulia Satiaputra
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, Australia
| | - Kira H Wood
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, Australia
| | - Devina D Lakhiani
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, Australia
| | - Abate A Bashaw
- Melanoma Discovery Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, Australia
| | - Lisa M Nilsson
- Melanoma Discovery Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, Australia
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical Sciences, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ji Li
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, Australia
| | - Edward R Bastow
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, Australia
| | - Anna Johansson-Percival
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, Australia
| | - Elena Denisenko
- Systems Biology and Genomics Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, Australia
| | - Alistair Rr Forrest
- Systems Biology and Genomics Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, Australia
| | | | - Rafael Carretero
- DKFZ-Bayer Immunotherapeutic Lab, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Jonas A Nilsson
- Melanoma Discovery Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, Australia
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical Sciences, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Gabriel Yf Lee
- St. John of God Subiaco Hospital and School of Surgery, The University of Western Australia, Perth, Western Australia, Australia
| | - Ruth Ganss
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
2
|
Zhou Y, Tao L, Qiu J, Xu J, Yang X, Zhang Y, Tian X, Guan X, Cen X, Zhao Y. Tumor biomarkers for diagnosis, prognosis and targeted therapy. Signal Transduct Target Ther 2024; 9:132. [PMID: 38763973 PMCID: PMC11102923 DOI: 10.1038/s41392-024-01823-2] [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: 06/05/2023] [Revised: 03/07/2024] [Accepted: 04/02/2024] [Indexed: 05/21/2024] Open
Abstract
Tumor biomarkers, the substances which are produced by tumors or the body's responses to tumors during tumorigenesis and progression, have been demonstrated to possess critical and encouraging value in screening and early diagnosis, prognosis prediction, recurrence detection, and therapeutic efficacy monitoring of cancers. Over the past decades, continuous progress has been made in exploring and discovering novel, sensitive, specific, and accurate tumor biomarkers, which has significantly promoted personalized medicine and improved the outcomes of cancer patients, especially advances in molecular biology technologies developed for the detection of tumor biomarkers. Herein, we summarize the discovery and development of tumor biomarkers, including the history of tumor biomarkers, the conventional and innovative technologies used for biomarker discovery and detection, the classification of tumor biomarkers based on tissue origins, and the application of tumor biomarkers in clinical cancer management. In particular, we highlight the recent advancements in biomarker-based anticancer-targeted therapies which are emerging as breakthroughs and promising cancer therapeutic strategies. We also discuss limitations and challenges that need to be addressed and provide insights and perspectives to turn challenges into opportunities in this field. Collectively, the discovery and application of multiple tumor biomarkers emphasized in this review may provide guidance on improved precision medicine, broaden horizons in future research directions, and expedite the clinical classification of cancer patients according to their molecular biomarkers rather than organs of origin.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lei Tao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiahao Qiu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Xu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinyu Yang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yu Zhang
- West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
- School of Medicine, Tibet University, Lhasa, 850000, China
| | - Xinyu Tian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xinqi Guan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaobo Cen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yinglan Zhao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
3
|
Pathak A, Pal AK, Roy S, Nandave M, Jain K. Role of Angiogenesis and Its Biomarkers in Development of Targeted Tumor Therapies. Stem Cells Int 2024; 2024:9077926. [PMID: 38213742 PMCID: PMC10783989 DOI: 10.1155/2024/9077926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/21/2023] [Accepted: 12/04/2023] [Indexed: 01/13/2024] Open
Abstract
Angiogenesis plays a significant role in the human body, from wound healing to tumor progression. "Angiogenic switch" indicates a time-restricted event where the imbalance between pro- and antiangiogenic factors results in the transition from prevascular hyperplasia to outgrowing vascularized tumor, which eventually leads to the malignant cancer progression. In the last decade, molecular players, i.e., angiogenic biomarkers and underlying molecular pathways involved in tumorigenesis, have been intensely investigated. Disrupting the initiation and halting the progression of angiogenesis by targeting these biomarkers and molecular pathways has been considered as a potential treatment approach for tumor angiogenesis. This review discusses the currently known biomarkers and available antiangiogenic therapies in cancer, i.e., monoclonal antibodies, aptamers, small molecular inhibitors, miRNAs, siRNAs, angiostatin, endostatin, and melatonin analogues, either approved by the U.S. Food and Drug Administration or currently under clinical and preclinical investigations.
Collapse
Affiliation(s)
- Anchal Pathak
- Drug Delivery and Nanomedicine Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, India
| | - Ajay Kumar Pal
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 110017, India
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | - Mukesh Nandave
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 110017, India
| | - Keerti Jain
- Drug Delivery and Nanomedicine Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, India
| |
Collapse
|
4
|
Pandey P, Khan F, Upadhyay TK, Seungjoon M, Park MN, Kim B. New insights about the PDGF/PDGFR signaling pathway as a promising target to develop cancer therapeutic strategies. Biomed Pharmacother 2023; 161:114491. [PMID: 37002577 DOI: 10.1016/j.biopha.2023.114491] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 02/20/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023] Open
Abstract
Numerous cancers express platelet-derived growth factors (PDGFs) and PDGF receptors (PDGFRs). By directly stimulating tumour cells in an autocrine manner or by stimulating tumour stromal cells in a paracrine manner, the platelet-derived growth factor (PDGF)/platelet-derived growth factor receptor (PDGFR) pathway is crucial in the growth and spread of several cancers. To combat hypoxia in the tumour microenvironment, it encourages angiogenesis. A growing body of experimental data shows that PDGFs target malignant cells, vascular cells, and stromal cells to modulate tumour growth, metastasis, and the tumour microenvironment. To combat medication resistance and enhance patient outcomes in cancers, targeting the PDGF/PDGFR pathway is a viable therapeutic approach. There have been reports of anomalies in the PDGF pathway, including the gain of function point mutations, activating chromosomal translocations, or overexpression or amplification of PDGF receptors (PDGFRs). As a result, it has been shown that targeting the PDGF/PDGFR signaling pathway is an effective method for treating cancer. As a result, this study will concentrate on the regulation of the PDGF/PDGFR signaling system, in particular the current methods and inhibitors used in cancer treatment, as well as the associated therapeutic advantages and side effects.
Collapse
Affiliation(s)
- Pratibha Pandey
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, UP, India
| | - Fahad Khan
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, UP, India.
| | - Tarun Kumar Upadhyay
- Department of Biotechnology, Parul Institute of Applied Sciences and Centre of Research for Development, Parul University, Vadodara 391760, India
| | - Moon Seungjoon
- Chansol Hospital of Korean Medicine, 290, Buheung-ro, Bupyeong-gu, Incheon 21390, Republic of Korea; Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea; Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea; Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
5
|
Zou X, Tang XY, Qu ZY, Sun ZW, Ji CF, Li YJ, Guo SD. Targeting the PDGF/PDGFR signaling pathway for cancer therapy: A review. Int J Biol Macromol 2022; 202:539-557. [PMID: 35074329 DOI: 10.1016/j.ijbiomac.2022.01.113] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 02/06/2023]
Abstract
Platelet-derived growth factors (PDGFs) and PDGF receptors (PDGFRs) are expressed in a variety of tumors. Activation of the PDGF/PDGFR signaling pathway is associated with cancer proliferation, metastasis, invasion, and angiogenesis through modulating multiple downstream pathways, including phosphatidylinositol 3 kinase/protein kinase B pathway and mitogen-activated protein kinase/extracellular signal-regulated kinase pathway. Therefore, targeting PDGF/PDGFR signaling pathway has been demonstrated to be an effective strategy for cancer therapy, and accordingly, some great progress has been made in this field in the past few decades. This review will focus on the PDGF isoforms and their binding with the related PDGFRs, the PDGF/PDGFR signaling and regulation, and especially present strategies and inhibitors developed for cancer therapy, and the related clinical benefits and side effects.
Collapse
Affiliation(s)
- Xiang Zou
- Engineering Research Center of Natural Antineoplastic Drugs, Ministry of Education, Harbin University of Commerce, 150076, China
| | - Xi-Yu Tang
- Engineering Research Center of Natural Antineoplastic Drugs, Ministry of Education, Harbin University of Commerce, 150076, China
| | - Zhong-Yuan Qu
- School of Pharmacy, Harbin University of Commerce, Harbin 150076, China.
| | - Zhi-Wei Sun
- School of Pharmacy, Harbin University of Commerce, Harbin 150076, China
| | - Chen-Feng Ji
- Engineering Research Center of Natural Antineoplastic Drugs, Ministry of Education, Harbin University of Commerce, 150076, China
| | - Yan-Jie Li
- Institute of lipid metabolism and Atherosclerosis, School of Pharmacy, Weifang Medical University, Weifang 261053, China.
| | - Shou-Dong Guo
- Engineering Research Center of Natural Antineoplastic Drugs, Ministry of Education, Harbin University of Commerce, 150076, China; School of Pharmacy, Harbin University of Commerce, Harbin 150076, China; Institute of lipid metabolism and Atherosclerosis, School of Pharmacy, Weifang Medical University, Weifang 261053, China.
| |
Collapse
|
6
|
Cui X, Jia H, Xin H, Zhang L, Chen S, Xia S, Li X, Xu W, Chen X, Feng Y, Wei X, Yu H, Wang Y, Zhan Y, Zhu X, Zhang X. A Novel Bispecific Antibody Targeting PD-L1 and VEGF With Combined Anti-Tumor Activities. Front Immunol 2021; 12:778978. [PMID: 34925354 PMCID: PMC8678608 DOI: 10.3389/fimmu.2021.778978] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/12/2021] [Indexed: 12/25/2022] Open
Abstract
Therapeutic monoclonal antibodies (mAbs) blocking immune checkpoints have been mainly used as monotherapy. Recently, combination therapy targeting multiple immune checkpoints has recently been explored to increase anti-cancer efficacy. Particularly, a single molecule targeting more than one checkpoints has been investigated. As dual blocking of PD-1/PD-L1 and VEGF/VEGFR has demonstrated synergism in anti-tumor activities, we developed a novel bispecific antibody, termed HB0025, which is formed via fusing the domain 2 of vascular endothelial growth factor receptor 1 (VEGFR1D2) and anti-PD-L1 mAb by using mAb-Trap technology. HB0025 almost completely retains the binding affinities and the biological activities in-vitro when compared with the parent anti-PD-L1 mAb or VEGFR1D2 fusion protein. Preclinical data demonstrated that HB0025 was more effective in inhibiting cancer growth than anti PD-L1 mAb or VEGFR1D2 fusion protein. Thus, our bispecific antibody may bring about greater clinical benefits and broader indications.
Collapse
Affiliation(s)
- Xiaopei Cui
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China.,Huabo Biopharma, Member of Zhejiang Huahai Pharmaceutical, Shanghai, China
| | - Huifeng Jia
- Huabo Biopharma, Member of Zhejiang Huahai Pharmaceutical, Shanghai, China
| | - Hong Xin
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Lei Zhang
- Huaota Biopharma, Member of Zhejiang Huahai Pharmaceutical, Shanghai, China
| | - Shi Chen
- Huabo Biopharma, Member of Zhejiang Huahai Pharmaceutical, Shanghai, China
| | - Simin Xia
- Huabo Biopharma, Member of Zhejiang Huahai Pharmaceutical, Shanghai, China
| | - Xue Li
- Huabo Biopharma, Member of Zhejiang Huahai Pharmaceutical, Shanghai, China
| | - Wei Xu
- Huabo Biopharma, Member of Zhejiang Huahai Pharmaceutical, Shanghai, China
| | - Xiaofang Chen
- Huabo Biopharma, Member of Zhejiang Huahai Pharmaceutical, Shanghai, China
| | - Yujie Feng
- Huabo Biopharma, Member of Zhejiang Huahai Pharmaceutical, Shanghai, China
| | - Xiaoyue Wei
- Huabo Biopharma, Member of Zhejiang Huahai Pharmaceutical, Shanghai, China
| | - Haijia Yu
- Huabo Biopharma, Member of Zhejiang Huahai Pharmaceutical, Shanghai, China
| | - Yanting Wang
- Huabo Biopharma, Member of Zhejiang Huahai Pharmaceutical, Shanghai, China
| | - Yifan Zhan
- Huabo Biopharma, Member of Zhejiang Huahai Pharmaceutical, Shanghai, China
| | - Xiangyang Zhu
- Huabo Biopharma, Member of Zhejiang Huahai Pharmaceutical, Shanghai, China.,Huaota Biopharma, Member of Zhejiang Huahai Pharmaceutical, Shanghai, China
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
7
|
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: 47] [Impact Index Per Article: 11.8] [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
|
8
|
Hao H, Zhang M, Zhang G, Zhu H, Bao W. [Intervention of Vascular Dysplasia Caused by Different Mechanisms - Anlotinib for Right Lung Squamous Cell Carcinoma Combined with Thromboangiitis Obliterans: A Case Report and Literature Review]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2020; 23:60-64. [PMID: 31948540 PMCID: PMC7007391 DOI: 10.3779/j.issn.1009-3419.2020.01.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND To date, there is no effective treatment for thromboangiitis obliterans (TAO). Anlotinib, as a third-line therapy, is recommended for patients with refractory advanced non-small cell lung cancer (NSCLC). We presented a case report of a patient suffering from right lung squamous cell carcinoma combined with thromboangiitis obliterans, and analyzed the treatment dilemma, which provided a new idea for the treatment of these two diseases. METHODS A patient of right lung squamous cell carcinoma complicated with TAO was admitted to the department of respiratory and critical care medicine of the Shanghai General Hospital in August 2018. The diagnosis and treatment was retrospectively analyzed, and the literature was reviewed. RESULTS The 73-year-old male patient complained of cough and sputum for 5 months and was diagnosed with NSCLC (T4N2M0, stage IIIb, performance status score 2) in right upper lung by tracheoscopy biopsy. Pigmentation in both lower extremities accompanied by weakened pulse of dorsal foot artery was confirmed. He had a history of smoking, and suspected vascular intermittent claudication and wandering phlebitis for more than one year. Ultrasound indicated multiple arterial occlusion in both upper and lower extremities and deep venous thrombosis in lower extremities. TAO was diagnosed. Peripherally inserted central catheter (PICC) implantation and intravenous infusion post implantation failed and he could not receive chemotherapy. Vascular endothelial growth factor (VEGF) signal pathway dysfunction is also involved in TAO. Anlotinib (12 mg qd po) was selected for treatment NSCLC and TAO, accordingly. He had partial response (PR) and the cancer kept stable for 14 months. At the same time, TAO improved. CONCLUSIONS Anlotinib effectively controlled the growth of NSCLC and improved TAO related symptoms. Anlotinib maybe normalize disordered growth of blood vessels through the VEGF signaling pathway, rather than simply inhibiting angiogenesis.
Collapse
Affiliation(s)
- Huijuan Hao
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Min Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Guoqing Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Hui Zhu
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Wuping Bao
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| |
Collapse
|
9
|
Fu Z, Xiang J. Aptamers, the Nucleic Acid Antibodies, in Cancer Therapy. Int J Mol Sci 2020; 21:ijms21082793. [PMID: 32316469 PMCID: PMC7215806 DOI: 10.3390/ijms21082793] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/09/2020] [Accepted: 04/15/2020] [Indexed: 02/06/2023] Open
Abstract
The arrival of the monoclonal antibody (mAb) technology in the 1970s brought with it the hope of conquering cancers to the medical community. However, mAbs, on the whole, did not achieve the expected wonder in cancer therapy although they do have demonstrated successfulness in the treatment of a few types of cancers. In 1990, another technology of making biomolecules capable of specific binding appeared. This technique, systematic evolution of ligands by exponential enrichment (SELEX), can make aptamers, single-stranded DNAs or RNAs that bind targets with high specificity and affinity. Aptamers have some advantages over mAbs in therapeutic uses particularly because they have little or no immunogenicity, which means the feasibility of repeated use and fewer side effects. In this review, the general properties of the aptamer, the advantages and limitations of aptamers, the principle and procedure of aptamer production with SELEX, particularly the undergoing studies in aptamers for cancer therapy, and selected anticancer aptamers that have entered clinical trials or are under active investigations are summarized.
Collapse
Affiliation(s)
- Zhaoying Fu
- Department of Biochemistry and Molecular Biology, College of Medicine, Yanan University, Yanan 716000, China
- Correspondence: (Z.F.); (J.X.)
| | - Jim Xiang
- Division of Oncology, University of Saskatchewan, Saskatoon, SA S7N 4H4, Canada
- Correspondence: (Z.F.); (J.X.)
| |
Collapse
|
10
|
Ganss R. Tumour vessel remodelling: new opportunities in cancer treatment. VASCULAR BIOLOGY 2020; 2:R35-R43. [PMID: 32923973 PMCID: PMC7439841 DOI: 10.1530/vb-19-0032] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 01/14/2020] [Indexed: 12/17/2022]
Abstract
Tumour growth critically depends on a supportive microenvironment, including the tumour vasculature. Tumour blood vessels are structurally abnormal and functionally anergic which limits drug access and immune responses in solid cancers. Thus, tumour vasculature has been considered an attractive therapeutic target for decades. However, with time, anti-angiogenic therapy has evolved from destruction to structural and functional rehabilitation as understanding of tumour vascular biology became more refined. Vessel remodelling or normalisation strategies which alleviate hypoxia are now coming of age having been shown to have profound effects on the tumour microenvironment. This includes improved tumour perfusion, release from immune suppression and lower metastasis rates. Nevertheless, clinical translation has been slow due to challenges such as the transient nature of current normalisation strategies, limited in vivo monitoring and the heterogeneity of primary and/or metastatic tumour environments, calling for more tailored approaches to vascular remodelling. Despite these setbacks, harnessing vascular plasticity provides unique opportunities for anti-cancer combination therapies in particular anti-angiogenic immunotherapy which are yet to reach their full potential.
Collapse
Affiliation(s)
- Ruth Ganss
- Vascular Biology and Stromal Targeting, Harry Perkins Institute of Medical Research, The University of Western Australia, Centre for Medical Research, Nedlands, Western Australia, Australia
| |
Collapse
|
11
|
Tsioumpekou M, Cunha SI, Ma H, Åhgren A, Cedervall J, Olsson AK, Heldin CH, Lennartsson J. Specific targeting of PDGFRβ in the stroma inhibits growth and angiogenesis in tumors with high PDGF-BB expression. Am J Cancer Res 2020; 10:1122-1135. [PMID: 31938055 PMCID: PMC6956815 DOI: 10.7150/thno.37851] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 10/01/2019] [Indexed: 01/12/2023] Open
Abstract
PDGF-BB/PDGFRβ signaling plays an important role during vascularization by mediating pericyte recruitment to the vasculature, promoting the integrity and function of vessels. Until now it has not been possible to assess the specific role of PDGFRβ signaling in tumor progression and angiogenesis due to lack of appropriate animal models and molecular tools. Methods: In the present study, we used a transgenic knock-in mouse strain carrying a silent mutation in the PDGFRβ ATP binding site that allows specific targeting of PDGFRβ using the compound 1-NaPP1. To evaluate the impact of selective PDGFRβ inhibition of stromal cells on tumor growth we investigated four tumor cell lines with no or low PDGFRβ expression, i.e. Lewis lung carcinoma (LLC), EO771 breast carcinoma, B16 melanoma and a version of B16 that had been engineered to overexpress PDGF-BB (B16/PDGF-BB). Results: We found that specific impairment of PDGFRβ kinase activity by 1-NaPP1 treatment efficiently suppressed growth in tumors with high expression of PDGF-BB, i.e. LLC and B16/PDGF-BB, while the clinically used PDGFRβ kinase inhibitor imatinib did not suppress tumor growth. Notably, tumors with low levels of PDGF-BB, i.e. EO771 and B16, neither responded to 1-NaPP1 nor to imatinib treatment. Inhibition of PDGFRβ by either drug impaired tumor vascularization and also affected pericyte coverage; however, specific targeting of PDGFRβ by 1-NaPP1 resulted in a more pronounced decrease in vessel function with increased vessel apoptosis in high PDGF-BB expressing tumors, compared to treatment with imatinib. In vitro analysis of PDGFRβ ASKA mouse embryo fibroblasts and the mesenchymal progenitor cell line 10T1/2 revealed that PDGF-BB induced NG2 expression, consistent with the in vivo data. Conclusion: Specific targeting of PDGFRβ signaling significantly inhibits tumor progression and angiogenesis depending on PDGF-BB expression. Our data suggest that targeting PDGFRβ in the tumor stroma could have therapeutic value in patients with high tumor PDGF-BB expression.
Collapse
|
12
|
Kilvaer TK, Rakaee M, Hellevik T, Vik J, Petris LD, Donnem T, Strell C, Ostman A, Busund LTR, Martinez-Zubiaurre I. Differential prognostic impact of platelet-derived growth factor receptor expression in NSCLC. Sci Rep 2019; 9:10163. [PMID: 31308421 PMCID: PMC6629689 DOI: 10.1038/s41598-019-46510-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 06/28/2019] [Indexed: 12/25/2022] Open
Abstract
Preclinical evidence suggests that stromal expression of platelet-derived growth factor receptors (PDGFRs) stimulates tumor development and diminishes intratumoral drug uptake. In non-small cell lung cancer (NSCLC), the clinical relevance of stromal PDGFR expression remains uncertain. Tumor specimens from 553 patients with primary operable stage I-IIIB NSCLC was obtained and tissue micro-arrays (TMA) were constructed (Norwegian cohort). Immunohistochemistry (IHC) was used to evaluate the expression of PDGFRα and -β in stromal cells and to explore their impact on patient survival. Results were validated in a non-related cohort consisting of TMAs of 367 stage I (A and B) NSCLC patients (Swedish cohort). High stromal PDGFRα expression was an independent predictor of increased survival in the overall populations and SCC (squamous cell carcinoma) subgroups of both investigated cohorts. PDGFRβ was an independent predictor of poor survival in the overall Norwegian cohort and an independent predictor of increased survival in the ADC (adenocarcinoma) subgroup of the Swedish cohort. Tumors displaying the combination PDGFRα-low/PDGFRβ-high exhibited inferior survival according to increasing stage in the Norwegian cohort. This study confirms that high stromal expression of PDGFRα is a predictor of increased survival in NSCLC. Further exploration of the prognostic impact of PDGFRβ and the relationship between PDGFRα and -β is warranted.
Collapse
Affiliation(s)
- Thomas Karsten Kilvaer
- Department of Oncology, University Hospital of North Norway, Tromso, Norway. .,Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway.
| | - Mehrdad Rakaee
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway.,Institute of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
| | - Turid Hellevik
- Department of Oncology, University Hospital of North Norway, Tromso, Norway.,Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
| | - Jørg Vik
- Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
| | - Luigi De Petris
- Department of Oncology-Pathology Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Tom Donnem
- Department of Oncology, University Hospital of North Norway, Tromso, Norway.,Institute of Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
| | - Carina Strell
- Department of Oncology-Pathology Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Arne Ostman
- Department of Oncology-Pathology Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Lill-Tove Rasmussen Busund
- Institute of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway.,Department of Clinical Pathology, University Hospital of North Norway, Tromso, Norway
| | | |
Collapse
|
13
|
Zobniw CM, Trinh VA, Posey K, Somaiah N. Olaratumab in the management of advanced soft tissue sarcoma. J Oncol Pharm Pract 2018; 25:442-448. [PMID: 30032714 DOI: 10.1177/1078155218788135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Olaratumab, the first-in-class anti-PDGFRα monoclonal antibody, has been contingently approved in combination with doxorubicin to treat adult patients with advanced soft tissue sarcoma for improving progression-free and overall survival. Olaratumab-doxorubicin combination has tolerable safety profile, which mimics that of doxorubicin monotherapy, with the exception of infusion-related reactions. Survival data of an ongoing confirmatory phase 3 trial are forthcoming to ascertain the optimal role of this product in the management algorithm of advanced soft tissue sarcoma. Active research is ongoing to identify biomarkers predictive of clinical benefit to olaratumab, to expand its utility to the pediatric population, and to explore its safety and efficacy in combination with other active regimens.
Collapse
Affiliation(s)
- Chrystia M Zobniw
- 1 Division of Pharmacy, Pharmacy Clinical Programs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Van Anh Trinh
- 1 Division of Pharmacy, Pharmacy Clinical Programs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kristi Posey
- 2 Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Neeta Somaiah
- 2 Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
14
|
Seynhaeve ALB, Oostinga D, van Haperen R, Eilken HM, Adams S, Adams RH, Ten Hagen TLM. Spatiotemporal endothelial cell - pericyte association in tumors as shown by high resolution 4D intravital imaging. Sci Rep 2018; 8:9596. [PMID: 29941944 PMCID: PMC6018425 DOI: 10.1038/s41598-018-27943-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 06/12/2018] [Indexed: 12/21/2022] Open
Abstract
Endothelial cells and pericytes are integral cellular components of the vasculature with distinct interactive functionalities. To study dynamic interactions between these two cells we created two transgenic animal lines. A truncated eNOS (endothelial nitric oxide synthase) construct was used as a GFP tag for endothelial cell evaluation and an inducible Cre-lox recombination, under control of the Pdgfrb (platelet derived growth factor receptor beta) promoter, was created for pericyte assessment. Also, eNOStag-GFP animals were crossed with the already established Cspg4-DsRed mice expressing DsRed fluorescent protein in pericytes. For intravital imaging we used tumors implanted in the dorsal skinfold of these transgenic animals. This setup allowed us to study time and space dependent complexities, such as distribution, morphology, motility, and association between both vascular cell types in all angiogenetic stages, without the need for additional labeling. Moreover, as fluorescence was still clearly detectable after fixation, it is possible to perform comparative histology following intravital evaluation. These transgenic mouse lines form an excellent model to capture collective and individual cellular and subcellular endothelial cell – pericyte dynamics and will help answer key questions on the cellular and molecular relationship between these two cells.
Collapse
Affiliation(s)
- Ann L B Seynhaeve
- Laboratory Experimental Surgical Oncology, Department of Surgery, Erasmus MC, 3015CE, Rotterdam, The Netherlands.
| | - Douwe Oostinga
- Laboratory Experimental Surgical Oncology, Department of Surgery, Erasmus MC, 3015CE, Rotterdam, The Netherlands
| | - Rien van Haperen
- Department of Cell Biology, Erasmus MC, 3015CE, Rotterdam, The Netherlands
| | - Hanna M Eilken
- Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, and Faculty of Medicine, University of Münster, D-48149, Münster, Germany
| | - Susanne Adams
- Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, and Faculty of Medicine, University of Münster, D-48149, Münster, Germany
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, and Faculty of Medicine, University of Münster, D-48149, Münster, Germany
| | - Timo L M Ten Hagen
- Laboratory Experimental Surgical Oncology, Department of Surgery, Erasmus MC, 3015CE, Rotterdam, The Netherlands
| |
Collapse
|
15
|
Zhang B, Wang H, Jin K, Jiang T, Shen S, Luo Z, Tuo Y, Jiang X, Liu X, Hu Y, Pang Z. BQ123 selectively improved tumor perfusion and enhanced nanomedicine delivery for glioblastomas treatment. Pharmacol Res 2018; 132:211-219. [DOI: 10.1016/j.phrs.2017.12.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 12/15/2017] [Accepted: 12/19/2017] [Indexed: 02/06/2023]
|
16
|
Optimization of the tumor microenvironment and nanomedicine properties simultaneously to improve tumor therapy. Oncotarget 2018; 7:62607-62618. [PMID: 27566585 PMCID: PMC5308750 DOI: 10.18632/oncotarget.11546] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 08/10/2016] [Indexed: 01/04/2023] Open
Abstract
Effective delivery of nanomedicines to tumor tissues depends on both the tumor microenvironment and nanomedicine properties. Accordingly, tumor microenvironment modification or advanced design of nanomedicine was emerging to improve nanomedicine delivery to tumors. However, few studies have emphasized the necessity to optimize the tumor microenvironment and nanomedicine properties simultaneously to improve tumor treatment. In the present study, imatinib mesylate (IMA) was used to normalize the tumor microenvironment including platelet-derived growth factor receptor-β expression inhibition, tumor vessel normalization, and tumor perfusion improvement as demonstrated by immunofluorescence staining. In addition, the effect of tumor microenvironment normalization on tumor delivery of nanomedicines with different sizes was carefully investigated. It was shown that IMA treatment significantly reduced the accumulation of nanoparticles (NPs) around 110 nm but enhanced the accumulation of micelles around 23 nm by in vivo fluorescence imaging experiment. Furthermore, IMA treatment limited the distribution of NPs inside tumors but increased that of micelles with a more homogeneous pattern. Finally, the anti-tumor efficacy study displayed that IMA pretreatment could significantly increase the therapeutic effects of paclitaxel-loaded micelles. All-together, a new strategy to improve nanomedicine delivery to tumor was provided by optimizing both nanomedicine size and the tumor microenvironment simultaneously, and it will have great potential in clinics for tumor treatment.
Collapse
|
17
|
Heldin CH, Lennartsson J, Westermark B. Involvement of platelet-derived growth factor ligands and receptors in tumorigenesis. J Intern Med 2018; 283:16-44. [PMID: 28940884 DOI: 10.1111/joim.12690] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Platelet-derived growth factor (PDGF) isoforms and their receptors have important roles during embryogenesis, particularly in the development of various mesenchymal cell types in different organs. In the adult, PDGF stimulates wound healing and regulates tissue homeostasis. However, overactivity of PDGF signalling is associated with malignancies and other diseases characterized by excessive cell proliferation, such as fibrotic conditions and atherosclerosis. In certain tumours, genetic or epigenetic alterations of the genes for PDGF ligands and receptors drive tumour cell proliferation and survival. Examples include the rare skin tumour dermatofibrosarcoma protuberance, which is driven by autocrine PDGF stimulation due to translocation of a PDGF gene, and certain gastrointestinal stromal tumours and leukaemias, which are driven by constitute activation of PDGF receptors due to point mutations and formation of fusion proteins of the receptors, respectively. Moreover, PDGF stimulates cells in tumour stroma and promotes angiogenesis as well as the development of cancer-associated fibroblasts, both of which promote tumour progression. Inhibitors of PDGF signalling may thus be of clinical usefulness in the treatment of certain tumours.
Collapse
Affiliation(s)
- C-H Heldin
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.,Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - J Lennartsson
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.,Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - B Westermark
- Department of Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
18
|
Tumor angiogenesis and vascular normalization: alternative therapeutic targets. Angiogenesis 2017; 20:409-426. [PMID: 28660302 DOI: 10.1007/s10456-017-9562-9] [Citation(s) in RCA: 1003] [Impact Index Per Article: 125.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 06/21/2017] [Indexed: 12/27/2022]
Abstract
Tumor blood vessels are a key target for cancer therapeutic management. Tumor cells secrete high levels of pro-angiogenic factors which contribute to the creation of an abnormal vascular network characterized by disorganized, immature and permeable blood vessels, resulting in poorly perfused tumors. The hypoxic microenvironment created by impaired tumor perfusion can promote the selection of more invasive and aggressive tumor cells and can also impede the tumor-killing action of immune cells. Furthermore, abnormal tumor perfusion also reduces the diffusion of chemotherapeutic drugs and radiotherapy efficiency. To fight against this defective phenotype, the normalization of the tumor vasculature has emerged as a new therapeutic strategy. Vascular normalization, by restoring proper tumor perfusion and oxygenation, could limit tumor cell invasiveness and improve the effectiveness of anticancer treatments. In this review, we investigate the mechanisms involved in tumor angiogenesis and describe strategies used to achieve vascular normalization.
Collapse
|
19
|
A combination of low-dose bevacizumab and imatinib enhances vascular normalisation without inducing extracellular matrix deposition. Br J Cancer 2017; 116:600-608. [PMID: 28141797 PMCID: PMC5344294 DOI: 10.1038/bjc.2017.13] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 01/05/2017] [Indexed: 01/18/2023] Open
Abstract
Background: Vascular endothelial growth factor (VEGF)-targeting drugs normalise the tumour vasculature and improve access for chemotherapy. However, excessive VEGF inhibition fails to improve clinical outcome, and successive treatment cycles lead to incremental extracellular matrix (ECM) deposition, which limits perfusion and drug delivery. We show here, that low-dose VEGF inhibition augmented with PDGF-R inhibition leads to superior vascular normalisation without incremental ECM deposition thus maintaining access for therapy. Methods: Collagen IV expression was analysed in response to VEGF inhibition in liver metastasis of colorectal cancer (CRC) patients, in syngeneic (Panc02) and xenograft tumours of human colorectal cancer cells (LS174T). The xenograft tumours were treated with low (0.5 mg kg−1 body weight) or high (5 mg kg−1 body weight) doses of the anti-VEGF antibody bevacizumab with or without the tyrosine kinase inhibitor imatinib. Changes in tumour growth, and vascular parameters, including microvessel density, pericyte coverage, leakiness, hypoxia, perfusion, fraction of vessels with an open lumen, and type IV collagen deposition were compared. Results: ECM deposition was increased after standard VEGF inhibition in patients and tumour models. In contrast, treatment with low-dose bevacizumab and imatinib produced similar growth inhibition without inducing detrimental collagen IV deposition, leading to superior vascular normalisation, reduced leakiness, improved oxygenation, more open vessels that permit perfusion and access for therapy. Conclusions: Low-dose bevacizumab augmented by imatinib selects a mature, highly normalised and well perfused tumour vasculature without inducing incremental ECM deposition that normally limits the effectiveness of VEGF targeting drugs.
Collapse
|
20
|
Paulsson J, Rydén L, Strell C, Frings O, Tobin NP, Fornander T, Bergh J, Landberg G, Stål O, Östman A. High expression of stromal PDGFRβ is associated with reduced benefit of tamoxifen in breast cancer. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2016; 3:38-43. [PMID: 28138400 PMCID: PMC5259559 DOI: 10.1002/cjp2.56] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 07/06/2016] [Accepted: 07/12/2016] [Indexed: 12/20/2022]
Abstract
Cancer‐associated fibroblasts (CAFs) regulate tumour growth, metastasis and response to treatment. Recent studies indicate the existence of functionally distinct CAF subsets. Suggested mechanisms whereby CAFs can impact on treatment response include paracrine signalling affecting cancer cell drug sensitivity and effects on tumour drug uptake. PDGFRβ is an important regulator of fibroblasts. Experimental studies have linked PDGFRβ‐positive fibroblasts to metastasis and also to reduced tumour drug uptake. This study has investigated the potential role of PDGFRβ‐positive fibroblasts in response to adjuvant tamoxifen treatment of breast cancer. Analyses of two breast cancer collections from randomised studies analysing adjuvant tamoxifen treatment in early breast cancer demonstrated significant benefit of tamoxifen in the group with low stromal PDGFRβ, which was not observed in the group with high stromal PDGFRβ. In general terms these findings provide novel evidence, derived from analyses of randomised clinical studies, of response‐predictive capacity of a marker‐defined subset of CAFs and, more specifically, identify stromal PDGFRβ as a marker related to tamoxifen benefit in early breast cancer.
Collapse
Affiliation(s)
- Janna Paulsson
- Department of Oncology-Pathology Cancer Center Karolinska, Karolinska Institutet Stockholm Sweden
| | - Lisa Rydén
- Division of Surgery, Department of Clinical SciencesLund UniversityLundSweden; Department of SurgerySkåne University HospitalLundSweden
| | - Carina Strell
- Department of Oncology-Pathology Cancer Center Karolinska, Karolinska Institutet Stockholm Sweden
| | - Oliver Frings
- Department of Oncology-Pathology Cancer Center Karolinska, Karolinska Institutet Stockholm Sweden
| | - Nicholas P Tobin
- Department of Oncology-Pathology Cancer Center Karolinska, Karolinska Institutet Stockholm Sweden
| | - Tommy Fornander
- Department of Oncology-Pathology Cancer Center Karolinska, Karolinska Institutet Stockholm Sweden
| | - Jonas Bergh
- Department of Oncology-PathologyCancer Center Karolinska, Karolinska InstitutetStockholmSweden; Radiumhemmet, Karolinska University HospitalStockholmSweden
| | - Göran Landberg
- Department of Pathology Sahlgrenska Cancer Centre, University of Gothenburg Gothenburg Sweden
| | - Olle Stål
- Department of Clinical and Experimental Medicine Oncology, Linköping University Linköping Sweden
| | - Arne Östman
- Department of Oncology-Pathology Cancer Center Karolinska, Karolinska Institutet Stockholm Sweden
| |
Collapse
|
21
|
Immobilized rolling circle amplification on extended-gate field-effect transistors with integrated readout circuits for early detection of platelet-derived growth factor. Anal Bioanal Chem 2016; 408:4785-97. [PMID: 27137518 DOI: 10.1007/s00216-016-9568-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 04/07/2016] [Accepted: 04/15/2016] [Indexed: 01/13/2023]
Abstract
Detection of tumor-related proteins with high specificity and sensitivity is important for early diagnosis and prognosis of cancers. While protein sensors based on antibodies are not easy to keep for a long time, aptamers (single-stranded DNA) are found to be a good alternative for recognizing tumor-related protein specifically. This study investigates the feasibility of employing aptamers to recognize the platelet-derived growth factor (PDGF) specifically and subsequently triggering rolling circle amplification (RCA) of DNAs on extended-gate field-effect transistors (EGFETs) to enhance the sensitivity. The EGFETs are fabricated by the standard CMOS technology and integrated with readout circuits monolithically. The monolithic integration not only avoids the wiring complexity for a large sensor array but also enhances the sensor reliability and facilitates massive production for commercialization. With the RCA primers immobilized on the sensory surface, the protein signal is amplified as the elongation of DNA, allowing the EGFET to achieve a sensitivity of 8.8 pM, more than three orders better than that achieved by conventional EGFETs. Moreover, the responses of EGFETs are able to indicate quantitatively the reaction rates of RCA, facilitating the estimation on the protein concentration. Our experimental results demonstrate that immobilized RCA on EGFETs is a useful, label-free method for early diagnosis of diseases related to low-concentrated tumor makers (e.g., PDGF) for serum sample, as well as for monitoring the synthesis of various DNA nanostructures in real time. Graphical Abstract The tumor-related protein, PDGF, is detected by immobilizing rolling circle amplification on an EGFET with integrated readout circuit.
Collapse
|
22
|
Zhang B, Jiang T, She X, Shen S, Wang S, Deng J, Shi W, Mei H, Hu Y, Pang Z, Jiang X. Fibrin degradation by rtPA enhances the delivery of nanotherapeutics to A549 tumors in nude mice. Biomaterials 2016; 96:63-71. [PMID: 27149664 DOI: 10.1016/j.biomaterials.2016.04.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/13/2016] [Accepted: 04/14/2016] [Indexed: 11/26/2022]
Abstract
Effective drug delivery to a tumor depends on favorable blood perfusion within the tumor. As an important component of tumor extracellular matrix, fibrin is abundant near tumor vessels. Inspired by the distinct distribution pattern and vessel-dependent production of fibrin, we hypothesized that fibrin depletion in tumors decompresses tumor vessels to improve tumor blood perfusion and accordingly enhance drug delivery to tumors rich in vessels. In the present study, we attempted to employ a clinically used thrombolytic drug, recombinant tissue plasminogen activator (rtPA), to modulate fibrin deposition in tumors. We then combined this drug with a nanoparticle drug delivery system for tumor therapy. RtPA treatment (25 mg/kg/d i. p. administration for two weeks) successfully depleted fibrin deposition and enhanced blood perfusion within A549 tumor xenografts. Furthermore, rtPA treatment also improved the in vivo delivery of 115-nm nanoparticles to tumor tissues. Finally, rtPA combined with therapeutic agent-loaded nanoparticles resulted in the most effective shrinkage of A549 tumor xenografts compared with the control groups. Overall, the present study provides a new strategy to enhance the delivery of nanotherapeutics to tumors rich in vessels.
Collapse
Affiliation(s)
- Bo Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, 430022, PR China
| | - Ting Jiang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, 430022, PR China
| | - Xiaojian She
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, PR China
| | - Shun Shen
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, PR China
| | - Sheng Wang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, PR China
| | - Jun Deng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, 430022, PR China
| | - Wei Shi
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, 430022, PR China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, 430022, PR China; Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, PR China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, 430022, PR China; Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, PR China.
| | - Zhiqing Pang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, PR China.
| | - Xinguo Jiang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, PR China
| |
Collapse
|
23
|
Zippel N, Ding Y, Fleming I. A Modified Aortic Ring Assay to Assess Angiogenic Potential In Vitro. Methods Mol Biol 2016; 1430:205-19. [PMID: 27172956 DOI: 10.1007/978-1-4939-3628-1_14] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Angiogenesis, an integral part of many physiological and pathological processes, is a tightly regulated multistep process. Angiogenesis assays are used to clarify the molecular mechanisms and screen for pharmacological inhibitors. However, most in vitro angiogenesis models measure only one aspect of this process, whereas in vivo assays are complex and difficult to interpret. The ex vivo aortic ring model allows the study of many key features of angiogenesis, such as endothelial activation, branching, and remodeling as well as later steps such as pericyte acquisition. This model can be modified to include genetic manipulation and can be used to assess the pro- or anti-angiogenic effects of compounds in a relatively controlled system.
Collapse
Affiliation(s)
- Nina Zippel
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, Frankfurt am Main, Frankfurt, D-60590, Germany
| | - Yindi Ding
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, Frankfurt am Main, Frankfurt, D-60590, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, Frankfurt am Main, Frankfurt, D-60590, Germany.
| |
Collapse
|
24
|
Wagner SC, Ichim TE, Ma H, Szymanski J, Perez JA, Lopez J, Bogin V, Patel AN, Marincola FM, Kesari S. Cancer anti-angiogenesis vaccines: Is the tumor vasculature antigenically unique? J Transl Med 2015; 13:340. [PMID: 26510973 PMCID: PMC4625691 DOI: 10.1186/s12967-015-0688-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 10/03/2015] [Indexed: 12/19/2022] Open
Abstract
Angiogenesis is essential for the growth and metastasis of solid tumors. The tumor endothelium exists in a state of chronic activation and proliferation, fueled by the tumor milieu where angiogenic mediators are aberrantly over-expressed. Uncontrolled tumor growth, immune evasion, and therapeutic resistance are all driven by the dysregulated and constitutive angiogenesis occurring in the vasculature. Accordingly, great efforts have been dedicated toward identifying molecular signatures of this pathological angiogenesis in order to devise selective tumor endothelium targeting therapies while minimizing potential autoimmunity against physiologically normal endothelium. Vaccination with angiogenic antigens to generate cellular and/or humoral immunity against the tumor endothelium has proven to be a promising strategy for inhibiting or normalizing tumor angiogenesis and reducing cancer growth. Here we review tumor endothelium vaccines developed to date including active immunization strategies using specific tumor endothelium-associated antigens and whole endothelial cell-based vaccines designed to elicit immune responses against diverse target antigens. Among the novel therapeutic options, we describe a placenta-derived endothelial cell vaccine, ValloVax™, a polyvalent vaccine that is antigenically similar to proliferating tumor endothelium and is supported by pre-clinical studies to be safe and efficacious against several tumor types.
Collapse
Affiliation(s)
- Samuel C Wagner
- Batu Biologics Inc., Towne Center Drive, San Diego, CA, 92121, USA.
| | - Thomas E Ichim
- Batu Biologics Inc., Towne Center Drive, San Diego, CA, 92121, USA.
| | - Hong Ma
- Batu Biologics Inc., Towne Center Drive, San Diego, CA, 92121, USA.
| | - Julia Szymanski
- Batu Biologics Inc., Towne Center Drive, San Diego, CA, 92121, USA.
| | | | - Javier Lopez
- Pan Am Cancer Treatment Center, Tijuana, Mexico.
| | - Vladimir Bogin
- Batu Biologics Inc., Towne Center Drive, San Diego, CA, 92121, USA.
| | - Amit N Patel
- Department of Surgery, University of Utah, Salt Lake City, UT, USA.
| | | | | |
Collapse
|
25
|
Chin VT, Nagrial AM, Chou A, Biankin AV, Gill AJ, Timpson P, Pajic M. Rho-associated kinase signalling and the cancer microenvironment: novel biological implications and therapeutic opportunities. Expert Rev Mol Med 2015; 17:e17. [PMID: 26507949 PMCID: PMC4836205 DOI: 10.1017/erm.2015.17] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The Rho/ROCK pathway is involved in numerous pivotal cellular processes that have made it an area of intense study in cancer medicine, however, Rho-associated coiled-coil containing protein kinase (ROCK) inhibitors are yet to make an appearance in the clinical cancer setting. Their performance as an anti-cancer therapy has been varied in pre-clinical studies, however, they have been shown to be effective vasodilators in the treatment of hypertension and post-ischaemic stroke vasospasm. This review addresses the various roles the Rho/ROCK pathway plays in angiogenesis, tumour vascular tone and reciprocal feedback from the tumour microenvironment and explores the potential utility of ROCK inhibitors as effective vascular normalising agents. ROCK inhibitors may potentially enhance the delivery and efficacy of chemotherapy agents and improve the effectiveness of radiotherapy. As such, repurposing of these agents as adjuncts to standard treatments may significantly improve outcomes for patients with cancer. A deeper understanding of the controlled and dynamic regulation of the key components of the Rho pathway may lead to effective use of the Rho/ROCK inhibitors in the clinical management of cancer.
Collapse
Affiliation(s)
- Venessa T. Chin
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia
| | - Adnan M. Nagrial
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia
- The Department of Medical Oncology, Crown Princess Mary Cancer Centre, Westmead Hospital, NSW, Australia
| | - Angela Chou
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia
- Anatomical Pathology, Sydpath, St Vincent's Hospital, Sydney, Australia
| | - Andrew V. Biankin
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia
- Department of Surgery, Bankstown Hospital, Eldridge Road, Bankstown, Sydney, NSW 2200, Australia
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow, Scotland G61 1BD, UK
| | - Anthony J. Gill
- Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia
- University of Sydney, Sydney, NSW 2006, Australia
| | - Paul Timpson
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia
- Faculty of Medicine, St Vincent's Clinical School, University of NSW, Australia
| | - Marina Pajic
- The Kinghorn Cancer Centre, Cancer Division, Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia
- Faculty of Medicine, St Vincent's Clinical School, University of NSW, Australia
| |
Collapse
|
26
|
Falcon BL, Swearingen M, Gough WH, Lee L, Foreman R, Uhlik M, Hanson JC, Lee JA, McClure DB, Chintharlapalli S. An in vitro cord formation assay identifies unique vascular phenotypes associated with angiogenic growth factors. PLoS One 2014; 9:e106901. [PMID: 25210890 PMCID: PMC4161374 DOI: 10.1371/journal.pone.0106901] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 08/11/2014] [Indexed: 01/08/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) plays a dominant role in angiogenesis. While inhibitors of the VEGF pathway are approved for the treatment of a number of tumor types, the effectiveness is limited and evasive resistance is common. One mechanism of evasive resistance to inhibition of the VEGF pathway is upregulation of other pro-angiogenic factors such as fibroblast growth factor (FGF) and epidermal growth factor (EGF). Numerous in vitro assays examine angiogenesis, but many of these assays are performed in media or matrix with multiple growth factors or are driven by VEGF. In order to study angiogenesis driven by other growth factors, we developed a basal medium to use on a co-culture cord formation system of adipose derived stem cells (ADSCs) and endothelial colony forming cells (ECFCs). We found that cord formation driven by different angiogenic factors led to unique phenotypes that could be differentiated and combination studies indicate dominant phenotypes elicited by some growth factors. VEGF-driven cords were highly covered by smooth muscle actin, and bFGF-driven cords had thicker nodes, while EGF-driven cords were highly branched. Multiparametric analysis indicated that when combined EGF has a dominant phenotype. In addition, because this assay system is run in minimal medium, potential proangiogenic molecules can be screened. Using this assay we identified an inhibitor that promoted cord formation, which was translated into in vivo tumor models. Together this study illustrates the unique roles of multiple anti-angiogenic agents, which may lead to improvements in therapeutic angiogenesis efforts and better rational for anti-angiogenic therapy.
Collapse
Affiliation(s)
- Beverly L. Falcon
- Department of Cancer Angiogenesis, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana, United States of America
| | - Michelle Swearingen
- Department of Cancer Angiogenesis, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana, United States of America
| | - Wendy H. Gough
- Department of Quantitative Biology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana, United States of America
| | - Linda Lee
- Department of Cancer Angiogenesis, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana, United States of America
| | - Robert Foreman
- Department of In Vivo Pharmacology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana, United States of America
| | - Mark Uhlik
- Department of Cancer Angiogenesis, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana, United States of America
| | - Jeff C. Hanson
- Department of Informatics Capabilities, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana, United States of America
| | - Jonathan A. Lee
- Department of Quantitative Biology, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana, United States of America
| | - Don B. McClure
- Department of BioTDR, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana, United States of America
| | - Sudhakar Chintharlapalli
- Department of Cancer Angiogenesis, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, Indiana, United States of America
- * E-mail:
| |
Collapse
|
27
|
Gacche RN, Meshram RJ. Angiogenic factors as potential drug target: Efficacy and limitations of anti-angiogenic therapy. Biochim Biophys Acta Rev Cancer 2014; 1846:161-79. [DOI: 10.1016/j.bbcan.2014.05.002] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 05/05/2014] [Accepted: 05/07/2014] [Indexed: 12/17/2022]
|
28
|
Using real-time impedance-based assays to monitor the effects of fibroblast-derived media on the adhesion, proliferation, migration and invasion of colon cancer cells. Biosci Rep 2014; 34:BSR20140031. [PMID: 24935351 PMCID: PMC4114067 DOI: 10.1042/bsr20140031] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Increasing our knowledge of the mechanisms regulating cell proliferation, migration and invasion are central to understanding tumour progression and metastasis. The local tumour microenvironment contributes to the transformed phenotype in cancer by providing specific environmental cues that alter the cells behaviour and promotes metastasis. Fibroblasts have a strong association with cancer and in recent times there has been some emphasis in designing novel therapeutic strategies that alter fibroblast behaviour in the tumour microenvironment. Fibroblasts produce growth factors, chemokines and many of the proteins laid down in the ECM (extracellular matrix) that promote angiogenesis, inflammation and tumour progression. In this study, we use a label-free RTCA (real-time cell analysis) platform (xCELLigence) to investigate how media derived from human fibroblasts alters cancer cell behaviour. We used a series of complimentary and novel experimental approaches to show HCT116 cells adhere, proliferate and migrate significantly faster in the presence of media from human fibroblasts. As well as this, we used the xCELLigence CIM-plates system to show that HCT116 cells invade matrigel layers aggressively when migrating towards media derived from human fibroblasts. These data strongly suggest that fibroblasts have the ability to increase the migratory and invasive properties of HCT116 cells. This is the first study that provides real-time data on fibroblast-mediated migration and invasion kinetics of colon cancer cells.
Collapse
|
29
|
Mallela J, Ravi S, Jean Louis F, Mulaney B, Cheung M, Sree Garapati U, Chinnasamy V, Wang C, Nagaraj S, Mohapatra SS, Mohapatra S. Natriuretic peptide receptor A signaling regulates stem cell recruitment and angiogenesis: a model to study linkage between inflammation and tumorigenesis. Stem Cells 2014; 31:1321-9. [PMID: 23533187 DOI: 10.1002/stem.1376] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 02/09/2013] [Accepted: 02/15/2013] [Indexed: 12/21/2022]
Abstract
Natriuretic peptide receptor A (NPRA), the signaling receptor for the cardiac hormone, atrial natriuretic peptide (ANP), is expressed abundantly in inflamed/injured tissues and tumors. NPRA deficiency substantially decreases tissue inflammation and inhibits tumor growth. However, the precise mechanism of NPRA function and whether it links inflammation and tumorigenesis remains unknown. Since both injury repair and tumor growth require stem cell recruitment and angiogenesis, we examined the role of NPRA signaling in tumor angiogenesis as a model of tissue injury repair in this study. In in vitro cultures, aortas from NPRA-KO mice show significantly lower angiogenic response compared to wild-type counterparts. The NPRA antagonist that decreases NPRA expression, inhibits lipopolysaccharide-induced angiogenesis. The reduction in angiogenesis correlates with decreased expression of vascular endothelial growth factor and chemokine (C-X-C motif) receptor 4 (CXCR4) implicating a cell recruitment defect. To test whether NPRA regulates migration of cells to tumors, mesenchymal stem cells (MSCs) were administered i.v., and the results showed that MSCs fail to migrate to the tumor microenvironment in NPRA-KO mice. However, coimplanting tumor cells with MSCs increases angiogenesis and tumorigenesis in NPRA-KO mice, in part by promoting expression of CXCR4 and its ligand, stromal cell-derived factor 1α. Taken together, these results demonstrate that NPRA signaling regulates stem cell recruitment and angiogenesis leading to tumor growth. Thus, NPRA signaling provides a key linkage between inflammation and tumorigenesis, and NPRA may be a target for drug development against cancers and tissue injury repair.
Collapse
Affiliation(s)
- Jaya Mallela
- Department of Molecular Medicine, University of South Florida, Tampa, FL, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Harper J, Sainson RCA. Regulation of the anti-tumour immune response by cancer-associated fibroblasts. Semin Cancer Biol 2014; 25:69-77. [PMID: 24406209 DOI: 10.1016/j.semcancer.2013.12.005] [Citation(s) in RCA: 198] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 12/23/2013] [Accepted: 12/30/2013] [Indexed: 02/07/2023]
Abstract
The microenvironment of established tumours is often immunosuppressed, and this allows tumours to grow and disseminate without being eliminated by the patient's immune system. The recent FDA approval of immunotherapies such as ipilimumab and sipuleucel-T that directly activate the adaptive and innate immune responses has triggered interest in developing other novel anti-cancer approaches that modulate the immune system. Understanding how the different constituents of the tumour microenvironment influence the immune system is thus crucial and is expected to generate a plethora of factors that can be targeted to boost immunity and trigger long lasting anti-tumour efficacy. Cancer associated fibroblasts (CAFs) are a crucial component of the tumour microenvironment. Through secretion of multiple growth factors, cytokines and proteases, CAFs are known to be key effectors for tumour progression and can promote cancer cell growth, invasiveness and angiogenesis. However, recent publications have also linked CAF biology to innate and adaptive immune cell recruitment and regulation. Here, we review recent findings on how CAFs can influence the immune status of tumours through direct and indirect interaction with immune cells and other key components of the tumour microenvironment.
Collapse
Affiliation(s)
- James Harper
- MedImmune Ltd., Granta Park, Cambridge CB21 6GH, UK.
| | | |
Collapse
|
31
|
Cortez E, Roswall P, Pietras K. Functional subsets of mesenchymal cell types in the tumor microenvironment. Semin Cancer Biol 2014; 25:3-9. [PMID: 24412106 DOI: 10.1016/j.semcancer.2013.12.010] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 12/19/2013] [Accepted: 12/30/2013] [Indexed: 11/26/2022]
Abstract
In the field of tumor biology, increasing attention is now focused on the complex interactions between various constituent cell types within the tumor microenvironment as being functionally important for the etiology of the disease. The detailed description of tumor-promoting properties of cancer-associated fibroblasts, endothelial cells, pericytes, and immune cells, introduces novel potential drug targets for improved cancer treatments, as well as a rationale for exploring the tumor stroma as a previously unchartered source for prognostic or predictive biomarkers. However, recent work highlights the fact that cellular identity is perhaps too broadly defined and that subdivision of each cell type may reveal functionally distinct subsets of cells. Here, we will review our current understanding of the diversity of different subsets of mesenchymal cells, i.e., cancer-associated fibroblasts and pericytes, residing within the tumor parenchyma.
Collapse
Affiliation(s)
- Eliane Cortez
- Lund University, Department of Laboratory Medicine Lund, Division of Translational Cancer Research, Medicon Village, Building 404:A3, SE-223 81 Lund, Sweden
| | - Pernilla Roswall
- Karolinska Institutet, Department of Medical Biochemistry and Biophysics, Scheeles Väg 2, SE-171 77 Stockholm, Sweden
| | - Kristian Pietras
- Lund University, Department of Laboratory Medicine Lund, Division of Translational Cancer Research, Medicon Village, Building 404:A3, SE-223 81 Lund, Sweden; Karolinska Institutet, Department of Medical Biochemistry and Biophysics, Scheeles Väg 2, SE-171 77 Stockholm, Sweden.
| |
Collapse
|
32
|
Patel S, Ngounou Wetie AG, Darie CC, Clarkson BD. Cancer secretomes and their place in supplementing other hallmarks of cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 806:409-42. [PMID: 24952195 DOI: 10.1007/978-3-319-06068-2_20] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The secretome includes all macromolecules secreted by cells, in particular conditions at defined times, allowing cell-cell communication. Cancer cell secretomes that are altered compared to normal cells have shown significant potential for elucidating cancer biology. Proteins of secretomes are secreted by various secretory pathways and can be studied using different methods. Cancer secretomes seem to play an important role in known hallmarks of cancers such as excessive proliferation, reduced apoptosis, immune invasion, angioneogenesis, alteration in energy metabolism, and development of resistance against anti-cancer therapy [1, 2]. If a significant role of an altered secretome can be identified in cancer cells, using advanced mass spectrometry-based techniques, this may allow researchers to screen and characterize the secretome proteins involved in cancer progression and open up new opportunities to develop new therapies. We aim to elaborate upon recent advances in cancer cell secretome analysis using different proteomics techniques. In this review, we highlight the role of the altered secretome in contributing to already recognized and emerging hallmarks of cancer and we discuss new challenges in the field of secretome analysis.
Collapse
Affiliation(s)
- Sapan Patel
- Memorial Sloan Kettering Cancer Center, Molecular Pharmacology and Chemistry Program, 415 East 68th Street, New York, NY, 10065, USA
| | | | | | | |
Collapse
|
33
|
Heldin CH. Targeting the PDGF signaling pathway in tumor treatment. Cell Commun Signal 2013; 11:97. [PMID: 24359404 PMCID: PMC3878225 DOI: 10.1186/1478-811x-11-97] [Citation(s) in RCA: 369] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 12/11/2013] [Indexed: 01/15/2023] Open
Abstract
Platelet-derived growth factor (PDGF) isoforms and PDGF receptors have important functions in the regulation of growth and survival of certain cell types during embryonal development and e.g. tissue repair in the adult. Overactivity of PDGF receptor signaling, by overexpression or mutational events, may drive tumor cell growth. In addition, pericytes of the vasculature and fibroblasts and myofibroblasts of the stroma of solid tumors express PDGF receptors, and PDGF stimulation of such cells promotes tumorigenesis. Inhibition of PDGF receptor signaling has proven to useful for the treatment of patients with certain rare tumors. Whether treatment with PDGF/PDGF receptor antagonists will be beneficial for more common malignancies is the subject for ongoing studies.
Collapse
Affiliation(s)
- Carl-Henrik Heldin
- Ludwig Institute for Cancer Research, Science for life laboratory, Uppsala University, Box 595SE-751 24 Uppsala, Sweden.
| |
Collapse
|
34
|
Platelet-derived growth factor. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
35
|
Ehnman M, Östman A. Therapeutic targeting of platelet-derived growth factor receptors in solid tumors. Expert Opin Investig Drugs 2013; 23:211-26. [PMID: 24206431 DOI: 10.1517/13543784.2014.847086] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Genetic aberrations that are associated with platelet-derived growth factor receptor (PDGFR) activity are frequently found in glioblastomas (10 - 15%), dermatofibrosarcoma protuberans (≤ 100%) and gastrointestinal stromal tumors (5%). Sequencing studies have also identified mutations at lower frequency in common cancer types. Preclinical evidence further suggests tumor stimulatory roles of PDGFRs expressed by tumor stroma cells and indicates a deleterious effect of stromal PDGFRs on intratumoral drug uptake. AREAS COVERED This review summarizes the present understanding of PDGF signaling in solid tumors based on experimental studies and clinical findings. It also provides a discussion of selected ongoing efforts to develop novel cancer therapies involving PDGFR inhibition with tyrosine kinase inhibitors or PDGFR-targeting monoclonal antibodies. EXPERT OPINION An increased molecular understanding of response and resistance mechanisms will be essential for therapeutic advances in PDGFR-directed cancer therapy. Further developments rely on clinical studies where systematic analyses of target status in malignant cells and in cells of the tumor stroma are included. Studies with combination therapies will be facilitated by selective PDGFR inhibitors with reduced side effects. Finally, development of improved companion diagnostics is of critical importance for patient selection and monitoring of therapeutic effects.
Collapse
Affiliation(s)
- Monika Ehnman
- Karolinska Institutet, Department of Oncology-Pathology , SE-17177 Stockholm , Sweden
| | | |
Collapse
|
36
|
Leinster DA, Colom B, Whiteford JR, Ennis DP, Lockley M, McNeish IA, Aurrand-Lions M, Chavakis T, Imhof BA, Balkwill FR, Nourshargh S. Endothelial cell junctional adhesion molecule C plays a key role in the development of tumors in a murine model of ovarian cancer. FASEB J 2013; 27:4244-53. [PMID: 23825230 PMCID: PMC3819510 DOI: 10.1096/fj.13-230441] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 06/24/2013] [Indexed: 01/05/2023]
Abstract
Junctional adhesion molecule C (JAM-C) is a transmembrane protein with significant roles in regulation of endothelial cell (EC) functions, including immune cell recruitment and angiogenesis. As these responses are important in promoting tumor growth, the role of EC JAM-C in tumor development was investigated using the ID8 syngeneic model of ovarian cancer. Within 10-15 wk, intraperitoneally injected ID8 cells form multiple tumor deposits and ascites that resemble human high-grade serous ovarian cancer. Compared to wild-type mice, survival in this model was increased in EC JAM-C knockouts (KOs; 88 vs. 96 d, P=0.04) and reduced in EC JAM-C transgenics (88 vs. 78.5 d, P=0.03), mice deficient in or overexpressing EC JAM-C, respectively. While tumor growth was significantly reduced in EC JAM-C KOs (87% inhibition at 10 wk, P<0.0005), this was not associated with alterations in tumor vessel density or immune cell infiltration. However, tumor microvessels from EC JAM-C-deficient mice exhibited reduced pericyte coverage and increased vascular leakage, suggesting a role for EC JAM-C in the development of functional tumor vessels. These findings provide evidence for a role for EC JAM-C in tumor growth and aggressiveness as well as recruitment of pericytes to newly formed blood vessels in a model of ovarian cancer.
Collapse
Affiliation(s)
- David A Leinster
- 2Centre for Microvascular Research, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M6BQ, UK.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Falcon BL, Stewart J, Ezell S, Hanson J, Wijsman J, Ye X, Westin E, Donoho G, Credille K, Uhlik MT. High-content multiplexed tissue imaging and quantification for cancer drug discovery. Drug Discov Today 2013; 18:510-22. [DOI: 10.1016/j.drudis.2012.08.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 07/11/2012] [Accepted: 08/23/2012] [Indexed: 01/01/2023]
|
38
|
Prognostic Significance in Breast Cancer of a Gene Signature Capturing Stromal PDGF Signaling. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:2037-47. [DOI: 10.1016/j.ajpath.2013.02.018] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 01/10/2013] [Accepted: 02/07/2013] [Indexed: 12/20/2022]
|
39
|
Carcinoma-associated fibroblasts are a promising therapeutic target. Cancers (Basel) 2013; 5:149-69. [PMID: 24216702 PMCID: PMC3730310 DOI: 10.3390/cancers5010149] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 01/21/2013] [Accepted: 01/23/2013] [Indexed: 12/21/2022] Open
Abstract
Human carcinomas frequently exhibit significant stromal reactions such as the so-called "desmoplastic stroma" or "reactive stroma", which is characterised by the existence of large numbers of stromal cells and extracellular matrix proteins. Carcinoma-associated fibroblasts (CAFs), which are rich in activated fibroblast populations exemplified by myofibroblasts, are among the predominant cell types present within the tumour-associated stroma. Increased numbers of stromal myofibroblasts are often associated with high-grade malignancies with poor prognoses in humans. CAF myofibroblasts possess abilities to promote primary tumour development, growth and progression by stimulating the processes of neoangiogenesis as well as tumour cell proliferation, survival, migration and invasion. Moreover, it has been demonstrated that CAFs serve as a niche supporting the metastatic colonisation of disseminated carcinoma cells in distant organs. Their contribution to primary and secondary malignancies makes these fibroblasts a potential therapeutic target and they also appear to be relevant to the development of drug resistance and tumour recurrence. This review summarises our current knowledge of tumour-promoting CAFs and discusses the therapeutic feasibility of targeting these cells as well as disrupting heterotypic interactions with other cell types in tumours that may improve the efficacy of current anti-tumour therapies.
Collapse
|
40
|
Abstract
Human carcinomas frequently exhibit significant stromal reactions such as the so-called "desmoplastic stroma" or "reactive stroma", which is characterised by the existence of large numbers of stromal cells and extracellular matrix proteins. Carcinoma-associated fibroblasts (CAFs), which are rich in activated fibroblast populations exemplified by myofibroblasts, are among the predominant cell types present within the tumour-associated stroma. Increased numbers of stromal myofibroblasts are often associated with high-grade malignancies with poor prognoses in humans. CAF myofibroblasts possess abilities to promote primary tumour development, growth and progression by stimulating the processes of neoangiogenesis as well as tumour cell proliferation, survival, migration and invasion. Moreover, it has been demonstrated that CAFs serve as a niche supporting the metastatic colonisation of disseminated carcinoma cells in distant organs. Their contribution to primary and secondary malignancies makes these fibroblasts a potential therapeutic target and they also appear to be relevant to the development of drug resistance and tumour recurrence. This review summarises our current knowledge of tumour-promoting CAFs and discusses the therapeutic feasibility of targeting these cells as well as disrupting heterotypic interactions with other cell types in tumours that may improve the efficacy of current anti-tumour therapies.
Collapse
|
41
|
Peña C, Céspedes MV, Lindh MB, Kiflemariam S, Mezheyeuski A, Edqvist PH, Hägglöf C, Birgisson H, Bojmar L, Jirström K, Sandström P, Olsson E, Veerla S, Gallardo A, Sjöblom T, Chang ACM, Reddel RR, Mangues R, Augsten M, Ostman A. STC1 expression by cancer-associated fibroblasts drives metastasis of colorectal cancer. Cancer Res 2012; 73:1287-97. [PMID: 23243022 DOI: 10.1158/0008-5472.can-12-1875] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Platelet-derived growth factor (PDGF) receptor signaling is a major functional determinant of cancer-associated fibroblasts (CAF). Elevated expression of PDGF receptors on stromal CAFs is associated with metastasis and poor prognosis, but mechanism(s) that underlie these connections are not understood. Here, we report the identification of the secreted glycoprotein stanniocalcin-1 (STC1) as a mediator of metastasis by PDGF receptor function in the setting of colorectal cancer. PDGF-stimulated fibroblasts increased migration and invasion of cocultured colorectal cancer cells in an STC1-dependent manner. Analyses of human colorectal cancers revealed significant associations between stromal PDGF receptor and STC1 expression. In an orthotopic mouse model of colorectal cancer, tumors formed in the presence of STC1-deficient fibroblasts displayed reduced intravasation of tumor cells along with fewer and smaller distant metastases formed. Our results reveal a mechanistic basis for understanding the contribution of PDGF-activated CAFs to cancer metastasis.
Collapse
Affiliation(s)
- Cristina Peña
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Deharvengt SJ, Tse D, Sideleva O, McGarry C, Gunn JR, Longnecker DS, Carriere C, Stan RV. PV1 down-regulation via shRNA inhibits the growth of pancreatic adenocarcinoma xenografts. J Cell Mol Med 2012; 16:2690-700. [PMID: 22568538 PMCID: PMC3435473 DOI: 10.1111/j.1582-4934.2012.01587.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 05/02/2012] [Indexed: 12/11/2022] Open
Abstract
PV1 is an endothelial-specific protein with structural roles in the formation of diaphragms in endothelial cells of normal vessels. PV1 is also highly expressed on endothelial cells of many solid tumours. On the basis of in vitro data, PV1 is thought to actively participate in angiogenesis. To test whether or not PV1 has a function in tumour angiogenesis and in tumour growth in vivo, we have treated pancreatic tumour-bearing mice by single-dose intratumoural delivery of lentiviruses encoding for two different shRNAs targeting murine PV1. We find that PV1 down-regulation by shRNAs inhibits the growth of established tumours derived from two different human pancreatic adenocarcinoma cell lines (AsPC-1 and BxPC-3). The effect observed is because of down-regulation of PV1 in the tumour endothelial cells of host origin, PV1 being specifically expressed in tumour vascular endothelial cells and not in cancer or other stromal cells. There are no differences in vascular density of tumours treated or not with PV1 shRNA, and gain and loss of function of PV1 in endothelial cells does not modify either their proliferation or migration, suggesting that tumour angiogenesis is not impaired. Together, our data argue that down-regulation of PV1 in tumour endothelial cells results in the inhibition of tumour growth via a mechanism different from inhibiting angiogenesis.
Collapse
MESH Headings
- Adenocarcinoma/blood supply
- Adenocarcinoma/genetics
- Adenocarcinoma/pathology
- Animals
- Base Sequence
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cell Line, Tumor
- Cell Movement/genetics
- Down-Regulation
- Drug Screening Assays, Antitumor
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Lentivirus/genetics
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Knockout
- Mice, Nude
- Molecular Sequence Data
- Neovascularization, Pathologic/genetics
- Pancreatic Neoplasms/blood supply
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/pathology
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- RNA, Small Interfering/pharmacology
- Stromal Cells/metabolism
- Stromal Cells/pathology
Collapse
Affiliation(s)
- Sophie J Deharvengt
- Departments of Pathology, Geisel School of Medicine at DartmouthLebanon, NH, USA
| | - Dan Tse
- Departments of Pathology, Geisel School of Medicine at DartmouthLebanon, NH, USA
| | - Olga Sideleva
- Departments of Pathology, Geisel School of Medicine at DartmouthLebanon, NH, USA
| | - Caitlin McGarry
- Departments of Pathology, Geisel School of Medicine at DartmouthLebanon, NH, USA
| | - Jason R Gunn
- Norris Cotton Cancer Center, Geisel School of Medicine at DartmouthLebanon, NH, USA
- Department of Engineering Sciences, Thayer School of EngineeringHanover, NH, USA
| | - Daniel S Longnecker
- Departments of Pathology, Geisel School of Medicine at DartmouthLebanon, NH, USA
- Norris Cotton Cancer Center, Geisel School of Medicine at DartmouthLebanon, NH, USA
| | - Catherine Carriere
- Medicine, Geisel School of Medicine at DartmouthLebanon, NH, USA
- Norris Cotton Cancer Center, Geisel School of Medicine at DartmouthLebanon, NH, USA
| | - Radu V Stan
- Departments of Pathology, Geisel School of Medicine at DartmouthLebanon, NH, USA
- Microbiology and Immunology, Geisel School of Medicine at DartmouthLebanon, NH, USA
- Heart and Vascular Research Center, Geisel School of Medicine at DartmouthLebanon, NH, USA
- Norris Cotton Cancer Center, Geisel School of Medicine at DartmouthLebanon, NH, USA
| |
Collapse
|
43
|
Marsh T, Pietras K, McAllister SS. Fibroblasts as architects of cancer pathogenesis. Biochim Biophys Acta Mol Basis Dis 2012; 1832:1070-8. [PMID: 23123598 DOI: 10.1016/j.bbadis.2012.10.013] [Citation(s) in RCA: 189] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 10/16/2012] [Accepted: 10/19/2012] [Indexed: 12/19/2022]
Abstract
Studies of epithelial cancers (i.e., carcinomas) traditionally focused on transformation of the epithelium (i.e., the cancer cells) and how aberrant signaling within the cancer cells modulates the surrounding tissue of origin. In more recent decades, the normal cells, blood vessels, molecules, and extracellular components that surround the tumor cells, collectively known as the "tumor microenvironment" or "stroma", have received increasing attention and are now thought to be key regulators of tumor initiation and progression. Of particular relevance to the work reviewed herein are the fibroblasts, which make up the major cell type within the microenvironment of most carcinomas. Due to their inherent heterogeneity, plasticity, and function, it is perhaps not surprising that fibroblasts are ideal modulators of normal and cancerous epithelium; however, these aspects also present challenges if we are to interrupt their tumor-supportive functions. Here, we review the current body of knowledge and the many questions that still remain about the special entity known as the cancer-associated fibroblast. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.
Collapse
Affiliation(s)
- Timothy Marsh
- Hematology Division, Brigham & Women's Hospital, Boston, MA 02115, USA
| | | | | |
Collapse
|
44
|
Bauman JE, Eaton KD, Wallace SG, Carr LL, Lee SJ, Jones DV, Arias-Pulido H, Cerilli LA, Martins RG. A Phase II study of pulse dose imatinib mesylate and weekly paclitaxel in patients aged 70 and over with advanced non-small cell lung cancer. BMC Cancer 2012; 12:449. [PMID: 23033932 PMCID: PMC3517479 DOI: 10.1186/1471-2407-12-449] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 10/02/2012] [Indexed: 11/28/2022] Open
Abstract
Background In non-small cell lung cancer (NSCLC), interstitial hypertension is a barrier to chemotherapy delivery, and is mediated by platelet derived growth factor receptor (PDGFR). Antagonizing PDGFR with imatinib may improve intra-tumoral delivery of paclitaxel, increasing response rate (RR). Methods This single-stage, open-label phase II study evaluated pulse dose imatinib and weekly paclitaxel in elderly patients with advanced NSCLC. Eligible patients were aged ≥ 70 with untreated, stage IIIB-IV NSCLC and ECOG performance status 0-2. Primary endpoint was RR. Secondary endpoints included median progression free and overall survival (PFS, OS) and correlatives of PDGFR pathway activation. Baseline Charlson Comorbidity Index (CCI) and Vulnerable Elder Survey-13 (VES-13) were correlated with outcomes. Results Thirty-four patients with median age 75 enrolled. Eleven of 29 (38%) were frail by VES-13 score. Overall RR was 11/34 (32%; 95% CI 17%-51%), meeting the primary endpoint. Median PFS and OS were 3.6 and 7.3 months, respectively. High tumoral PDGF-B expression predicted inferior PFS. Frail patients by VES-13 had significantly worse median PFS (3.2 vs. 4.5 months; p=0.02) and OS (4.8 vs. 12 months; p=0.02) than non-frail. Conclusions The combination of imatinib and paclitaxel had encouraging activity as measured by the primary endpoint of RR. However, PFS and OS were typical for elderly patients treated with single agent chemotherapy and the regimen is not recommended for further study. Adjunct imatinib did not overcome the established association of tumoral PDGF-B expression with inferior PFS. VES-13 was a powerful predictor of poor survival outcomes. Frailty should be further studied as a predictor of non-benefit from chemotherapy. Trial Registration ClinicalTrials.gov NCT01011075
Collapse
Affiliation(s)
- Julie E Bauman
- Department of Internal Medicine, Division of Hematology/Oncology and Biostatistics, University of New Mexico Cancer Center, Albuquerque, New Mexico, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abastado JP. The next challenge in cancer immunotherapy: controlling T-cell traffic to the tumor. Cancer Res 2012; 72:2159-61. [PMID: 22549945 DOI: 10.1158/0008-5472.can-11-3538] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
One of the steps that limits the efficacy of T-cell-based immunotherapy of cancer is T-cell access to the tumor. We recently showed that several chemotherapeutic drugs induce intratumoral expression of chemokines that attract effector T cells. Moreover, in a cohort of patients with melanoma who had been treated with dacarbazine, one of the most frequently used chemotherapies for metastatic melanoma, tumor response to the treatment correlated with intratumoral expression of T-cell-attracting chemokines and with T-cell infiltration. These findings reveal the possibility of developing novel systemic strategies aimed at improving T-cell homing to tumors. Such strategies, used alone or in combination with adoptive T-cell therapies or therapeutic cancer vaccines, may prove to be more efficient in prolonging patient survival.
Collapse
Affiliation(s)
- Jean-Pierre Abastado
- Singapore Immunology Network, Biomedical Sciences Institutes, A-STAR, Singapore.
| |
Collapse
|
46
|
Baker M, Robinson SD, Lechertier T, Barber PR, Tavora B, D'Amico G, Jones DT, Vojnovic B, Hodivala-Dilke K. Use of the mouse aortic ring assay to study angiogenesis. Nat Protoc 2011; 7:89-104. [PMID: 22193302 DOI: 10.1038/nprot.2011.435] [Citation(s) in RCA: 382] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Here we provide a protocol for quantitative three-dimensional ex vivo mouse aortic ring angiogenesis assays, in which developing microvessels undergo many key features of angiogenesis over a timescale similar to that observed in vivo. The aortic ring assay allows analysis of cellular proliferation, migration, tube formation, microvessel branching, perivascular recruitment and remodeling-all without the need for cellular dissociation-thus providing a more complete picture of angiogenic processes compared with traditional cell-based assays. Our protocol can be applied to aortic rings from embryonic stage E18 through to adulthood and can incorporate genetic manipulation, treatment with growth factors, drugs or siRNA. This robust assay allows assessment of the salient steps in angiogenesis and quantification of the developing microvessels, and it can be used to identify new modulators of angiogenesis. The assay takes 6-14 d to complete, depending on the age of the mice, treatments applied and whether immunostaining is performed.
Collapse
Affiliation(s)
- Marianne Baker
- Adhesion and Angiogenesis Laboratory, Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|