651
|
Abstract
Macrophage plasticity is an important feature of these innate immune cells. Macrophage phenotypes are divided into two categories, the classically activated macrophages (CAM, M1 phenotype) and the alternatively activated macrophages (AAM, M2 phenotype). M1 macrophages are commonly associated with the generation of proinflammatory cytokines, whereas M2 macrophages are anti-inflammatory and often associated with tumor progression and fibrosis development. Macrophages produce high levels of reactive oxygen species (ROS). Recent evidence suggests ROS can potentially regulate macrophage phenotype. In addition, macrophages phenotypes are closely related to their metabolic patterns, particularly fatty acid/cholesterol metabolism. In this review, we briefly summarize recent advances in macrophage polarization with special attention to their relevance to specific disease conditions and metabolic regulation of polarization. Understanding these metabolic switches can facilitate the development of targeted therapies for various diseases.
Collapse
Affiliation(s)
- Chao He
- Department of Medicine, University of Alabama at Birmingham, Alabama, USA
| | - A Brent Carter
- Department of Medicine, University of Alabama at Birmingham, Alabama, USA; Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Alabama, USA; Birmingham VAMC, Birmingham, Alabama, USA
| |
Collapse
|
652
|
Shemi A, Khvalevsky EZ, Gabai RM, Domb A, Barenholz Y. Multistep, effective drug distribution within solid tumors. Oncotarget 2015; 6:39564-77. [PMID: 26416413 PMCID: PMC4741846 DOI: 10.18632/oncotarget.5051] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 09/10/2015] [Indexed: 12/18/2022] Open
Abstract
The distribution of drugs within solid tumors presents a long-standing barrier for efficient cancer therapies. Tumors are highly resistant to diffusion, and the lack of blood and lymphatic flows suppresses convection. Prolonged, continuous intratumoral drug delivery from a miniature drug source offers an alternative to both systemic delivery and intratumoral injection. Presented here is a model of drug distribution from such a source, in a multistep process. At delivery onset the drug mainly affects the closest surroundings. Such 'priming' enables drug penetration to successive cell layers. Tumor 'void volume' (volume not occupied by cells) increases, facilitating lymphatic perfusion. The drug is then transported by hydraulic convection downstream along interstitial fluid pressure (IFP) gradients, away from the tumor core. After a week tumor cell death occurs throughout the entire tumor and IFP gradients are flattened. Then, the drug is transported mainly by 'mixing', powered by physiological bulk body movements. Steady state is achieved and the drug covers the entire tumor over several months. Supporting measurements are provided from the LODER system, releasing siRNA against mutated KRAS over months in pancreatic cancer in-vivo models. LODER was also successfully employed in a recent Phase 1/2 clinical trial with pancreatic cancer patients.
Collapse
Affiliation(s)
| | | | | | - Abraham Domb
- Faculty of Medicine - School of Pharmacy, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yechezkel Barenholz
- Membrane and Liposome Research Lab, Hebrew University Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
653
|
Grenga I, Kwilas AR, Donahue RN, Farsaci B, Hodge JW. Inhibition of the angiopoietin/Tie2 axis induces immunogenic modulation, which sensitizes human tumor cells to immune attack. J Immunother Cancer 2015; 3:52. [PMID: 26579226 PMCID: PMC4647578 DOI: 10.1186/s40425-015-0096-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/21/2015] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND The angiopoietin/Tie2 pathway is an attractive target for cancer therapy due to its well-known role in regulating angiogenesis. Trebananib, a recombinant peptide-Fc fusion protein, or peptibody, that binds to angiopoietin-1 (Ang1) and Ang2 to block their interaction with the Tie2 receptor, is under active clinical investigation. We investigated whether suppressing the angiopoietin/Tie2 pathway, using the preclinical version of Trebananib (mL4-3 and L1-7(N)), could increase the sensitivity of human tumor cells to immune-mediated lysis through immunogenic modulation, which would make Trebananib a promising candidate for combination with immunotherapy. METHODS We assessed human carcinoma cells for expression and activation of Ang1 and Ang2 and their receptor tyrosine kinase Tie2. In vitro, we exposed tumor cell lines expressing Tie2 to the peptibodies mL4-3 and L1-7(N), which inhibit the binding of Ang1 and Ang2 to Tie2, and assessed the cells for changes in viability, proliferation, surface phenotype, and sensitivity to attack by antigen-specific cytotoxic T lymphocytes (CTLs). RESULTS Suppression of the angiopoietin/Tie2 pathway using mL4-3 and L1-7(N) had no effect on the proliferation or viability of tumor cells. However, these inhibitors markedly altered tumor cell phenotype, rendering tumor cells significantly more sensitive to antigen-specific CTL killing. ICAM-1 was shown to be mechanistically involved in these inhibitors' ability to sensitize tumor cells to immune-mediated attack by functional blocking studies. CONCLUSION Our findings provide a rationale for the combination of agents targeting the angiopoietin/Tie2 pathway with cancer immunotherapies.
Collapse
Affiliation(s)
- Italia Grenga
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Center Drive, Room 8B13 MSC 1750, Bethesda, MD 20892 USA
| | - Anna R Kwilas
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Center Drive, Room 8B13 MSC 1750, Bethesda, MD 20892 USA
| | - Renee N Donahue
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Center Drive, Room 8B13 MSC 1750, Bethesda, MD 20892 USA
| | - Benedetto Farsaci
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Center Drive, Room 8B13 MSC 1750, Bethesda, MD 20892 USA
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Center Drive, Room 8B13 MSC 1750, Bethesda, MD 20892 USA
| |
Collapse
|
654
|
Role of vascular density and normalization in response to neoadjuvant bevacizumab and chemotherapy in breast cancer patients. Proc Natl Acad Sci U S A 2015; 112:14325-30. [PMID: 26578779 DOI: 10.1073/pnas.1518808112] [Citation(s) in RCA: 179] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Preoperative bevacizumab and chemotherapy may benefit a subset of breast cancer (BC) patients. To explore potential mechanisms of this benefit, we conducted a phase II study of neoadjuvant bevacizumab (single dose) followed by combined bevacizumab and adriamycin/cyclophosphamide/paclitaxel chemotherapy in HER2-negative BC. The regimen was well-tolerated and showed a higher rate of pathologic complete response (pCR) in triple-negative (TN)BC (11/21 patients or 52%, [95% confidence interval (CI): 30,74]) than in hormone receptor-positive (HR)BC [5/78 patients or 6% (95%CI: 2,14)]. Within the HRBCs, basal-like subtype was significantly associated with pCR (P = 0.007; Fisher exact test). We assessed interstitial fluid pressure (IFP) and tissue biopsies before and after bevacizumab monotherapy and circulating plasma biomarkers at baseline and before and after combination therapy. Bevacizumab alone lowered IFP, but to a smaller extent than previously observed in other tumor types. Pathologic response to therapy correlated with sVEGFR1 postbevacizumab alone in TNBC (Spearman correlation 0.610, P = 0.0033) and pretreatment microvascular density (MVD) in all patients (Spearman correlation 0.465, P = 0.0005). Moreover, increased pericyte-covered MVD, a marker of extent of vascular normalization, after bevacizumab monotherapy was associated with improved pathologic response to treatment, especially in patients with a high pretreatment MVD. These data suggest that bevacizumab prunes vessels while normalizing those remaining, and thus is beneficial only when sufficient numbers of vessels are initially present. This study implicates pretreatment MVD as a potential predictive biomarker of response to bevacizumab in BC and suggests that new therapies are needed to normalize vessels without pruning.
Collapse
|
655
|
Lin L, Chen YS, Yao YD, Chen JQ, Chen JN, Huang SY, Zeng YJ, Yao HR, Zeng SH, Fu YS, Song EW. CCL18 from tumor-associated macrophages promotes angiogenesis in breast cancer. Oncotarget 2015; 6:34758-73. [PMID: 26416449 PMCID: PMC4741488 DOI: 10.18632/oncotarget.5325] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 09/11/2015] [Indexed: 02/05/2023] Open
Abstract
The infiltration of tumor-associated macrophages (TAMs) is associated with extensive angiogenesis, which contributes to a poor prognosis in breast cancer. However, anti-angiogenic therapy with VEGF-specific monotherapy has been unsuccessful in treating breast cancer, and the molecular mechanisms associated with chemoresistance remain unclear. Here, we investigated whether CCL18, a chemokine produced by TAMs, can stimulate angiogenesis in breast cancer, as well as the underlying mechanisms. Double immunohistochemical staining for CCL18 and CD34/CD31/vWF was performed in 80 breast cancer samples to study the correlation between CCL18+ TAMs and microvascular density (MVD). Cocultures of TAMs with human umbilical vein endothelial cells (HUVECs) were used to model the inflammatory microenvironment, and CCL18-induced angiogenesis was evaluated both in vitro and in vivo. We demonstrated that CCL18+ TAM infiltration positively associated with MVD in breast cancer samples, which was correlated with tumor metastasis and poor prognosis. We confirmed, both in vitro and in vivo, that CCL18 and VEGF synergistically promoted endothelial cell migration and angiogenesis. Conversely, blocking CCL18 or VEGF with neutralizing antibodies synergistically inhibited the promigratory effects of TAMs. Silencing PITPNM3, a putative CCL18 receptor, on the surface of HUVECs abrogated CCL18-mediated promigration and the enhancement of HUVEC tube formation, independently of VEGFR signaling. Moreover, CCL18 exposure induced the endothelial-mesenchymal transformation and activated ERK and Akt/GSK-3β/Snail signaling in HUVECs, thereby contributing to its pro-angiogenic effects. In conclusion, our findings suggest that CCL18 released from TAMs promotes angiogenesis and tumor progression in breast cancer; thus, CCL18 may serve as a novel target for anti-angiogenic therapies.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Breast Neoplasms/blood supply
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/blood supply
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Chemokines, CC/metabolism
- Enzyme-Linked Immunosorbent Assay
- Female
- Flow Cytometry
- Fluorescent Antibody Technique
- Heterografts
- Humans
- Immunohistochemistry
- Macrophages/metabolism
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Neovascularization, Pathologic/metabolism
- RNA, Small Interfering
- Transfection
- Tumor Microenvironment/physiology
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Ling Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Department of Internal Medicine, the First Affiliated Hospital, Shantou University Medical College, Shantou 515041, P. R. China
| | - Yong-Song Chen
- Department of Internal Medicine, the First Affiliated Hospital, Shantou University Medical College, Shantou 515041, P. R. China
| | - Yan-Dan Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
| | - Jing-Qi Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
| | - Jia-Ning Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
| | - Song-Yin Huang
- Department of Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
| | - Yun-Jie Zeng
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
| | - He-Rui Yao
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
| | - Si-Hai Zeng
- Guangzhou Blood Center, Guangzhou 510120, P. R. China
| | - Yong-Shui Fu
- Guangzhou Blood Center, Guangzhou 510120, P. R. China
| | - Er-Wei Song
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
| |
Collapse
|
656
|
Ruksha TG, Aksenenko MB, Shvetsova YI. [Molecular and pathomorphological prognostic markers for melanoma: Current approaches and prospects]. Arkh Patol 2015; 77:71-77. [PMID: 26485783 DOI: 10.17116/patol201577471-77] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The incidence of melanoma demonstrates a persistent increasing tendency, which justifies the need to study and identify new prognostic markers for the development and course of this disease. The given paper shows current approaches to melanoma staging, including those to applying pathomorphological prognostic criteria, and discusses prospects for using the results of genomic and epigenomic studies of the carcinoma in clinical practice.
Collapse
Affiliation(s)
- T G Ruksha
- Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Ministry of Health of the Russia, Krasnoyarsk
| | - M B Aksenenko
- Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Ministry of Health of the Russia, Krasnoyarsk
| | - Yu I Shvetsova
- Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Ministry of Health of the Russia, Krasnoyarsk
| |
Collapse
|
657
|
Abstract
Harnessing the ability of the immune system to eradicate cancer has been a long-held goal of oncology. Work from the last two decades has finally brought immunotherapy into the forefront for cancer treatment, with demonstrable clinical success for aggressive tumors where other therapies had failed. In this review, we will discuss a range of therapies that are in different stages of clinical or preclinical development for companion animals with cancer, and which share the common objective of eliciting adaptive, anti-tumor immune responses. Even though challenges remain, manipulating the immune system holds significant promise to create durable responses and improve outcomes in companion animals with cancer. Furthermore, what we learn from this process will inform and accelerate development of comparable therapies for human cancer patients.
Collapse
|
658
|
Chen Y, Duda DG. Targeting immunosuppression after standard sorafenib treatment to facilitate immune checkpoint blockade in hepatocellular carcinoma - an auto-commentary on clinical potential and future development. Oncoimmunology 2015. [PMID: 26451297 DOI: 10.1080/2162402x.2015.1029703]] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2022] Open
Abstract
Immunotherapy has shown great promise to transform solid cancer treatment. The challenge is to optimally incorporate novel immunotherapeutics, such as immune checkpoint blockers, with standard therapies. This is well exemplified by multimodal therapies recently developed for liver cancer in which immunomodulation using CXCR4 inhibition prevented immunosuppression and enhanced sorafenib and anti-PD-1 therapeutic outcome.
Collapse
Affiliation(s)
- Yunching Chen
- Institute of Biomedical Engineering; National Tsing Hua University ; Hsinchu, Taiwan ; Steele Laboratories for Tumor Biology; Department of Radiation Oncology; Massachusetts General Hospital and Harvard Medical School ; Boston, MA USA
| | - Dan G Duda
- Steele Laboratories for Tumor Biology; Department of Radiation Oncology; Massachusetts General Hospital and Harvard Medical School ; Boston, MA USA
| |
Collapse
|
659
|
He Q, Guo S, Qian Z, Chen X. Development of individualized anti-metastasis strategies by engineering nanomedicines. Chem Soc Rev 2015; 44:6258-6286. [PMID: 26056688 PMCID: PMC4540626 DOI: 10.1039/c4cs00511b] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Metastasis is deadly and also tough to treat as it is much more complicated than the primary tumour. Anti-metastasis approaches available so far are far from being optimal. A variety of nanomedicine formulae provide a plethora of opportunities for developing new strategies and means for tackling metastasis. It should be noted that individualized anti-metastatic nanomedicines are different from common anti-cancer nanomedicines as they specifically target different populations of malignant cells. This review briefly introduces the features of the metastatic cascade, and proposes a series of nanomedicine-based anti-metastasis strategies aiming to block each metastatic step. Moreover, we also concisely introduce the advantages of several promising nanoparticle platforms and their potential for constructing state-of-the-art individualized anti-metastatic nanomedicines.
Collapse
Affiliation(s)
- Qianjun He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P. R. China.
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| | - Shengrong Guo
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P. R. China.
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
660
|
Binder PS, Prat J, Mutch DG. The future role of molecular staging in gynecologic cancer. Int J Gynaecol Obstet 2015; 131 Suppl 2:S127-31. [DOI: 10.1016/j.ijgo.2015.06.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
661
|
Lote H, Cafferkey C, Chau I. PD-1 and PD-L1 blockade in gastrointestinal malignancies. Cancer Treat Rev 2015; 41:893-903. [PMID: 26412280 DOI: 10.1016/j.ctrv.2015.09.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 09/15/2015] [Indexed: 01/10/2023]
Abstract
Immunotherapy represents a major breakthrough in cancer therapy in recent years. Immune-checkpoint blockade using PD-1 and PD-L1 antibodies appears to be one of the most promising immunotherapy approaches. Immunotherapy differs from conventional cancer treatment because of its ability to produce durable responses in some patients. In this review article, we explore the available evidence and summarise current clinical trials for PD-1 and PD-L1 blockade in gastrointestinal malignancies. The challenge now is to develop strategies to increase the efficacy of PD-1 and PD-L1 blockade in gastrointestinal cancer patients, such as combination therapy with chemotherapy, radiotherapy or other immunotherapy, along with validating biomarkers to select patients and personalise treatment.
Collapse
Affiliation(s)
- Hazel Lote
- Department of Medicine, Royal Marsden Hospital, London and Surrey, Sutton SM2 5PT, UK
| | - Catherine Cafferkey
- Department of Medicine, Royal Marsden Hospital, London and Surrey, Sutton SM2 5PT, UK
| | - Ian Chau
- Department of Medicine, Royal Marsden Hospital, London and Surrey, Sutton SM2 5PT, UK.
| |
Collapse
|
662
|
Coffelt SB, de Visser KE. Immune-mediated mechanisms influencing the efficacy of anticancer therapies. Trends Immunol 2015; 36:198-216. [PMID: 25857662 DOI: 10.1016/j.it.2015.02.006] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 02/18/2015] [Accepted: 02/18/2015] [Indexed: 01/26/2023]
Abstract
Conventional anticancer therapies, such as chemotherapy, radiotherapy, and targeted therapy, are designed to kill cancer cells. However, the efficacy of anticancer therapies is not only determined by their direct effects on cancer cells but also by off-target effects within the host immune system. Cytotoxic treatment regimens elicit several changes in immune-related parameters including the composition, phenotype, and function of immune cells. Here we discuss the impact of innate and adaptive immune cells on the success of anticancer therapy. In this context we examine the opportunities to exploit host immune responses to boost tumor clearing, and highlight the challenges facing the treatment of advanced metastatic disease.
Collapse
Affiliation(s)
- Seth B Coffelt
- Division of Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| | - Karin E de Visser
- Division of Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| |
Collapse
|
663
|
Abstract
Cancer immunotherapy was selected as the Breakthrough of the Year 2013 by the editors of Science, in part because of the successful treatment of refractory hematological malignancies with adoptive transfer of chimeric antigen receptor (CAR)-engineered T cells. Effective treatment of B cell leukemia may pave the road to future treatment of solid tumors, using similar approaches. The prostate expresses many unique proteins and, since the prostate gland is a dispensable organ, CAR T cells can potentially be used to target these tissue-specific antigens. However, the location and composition of prostate cancer metastases complicate the task of treating these tumors. It is therefore likely that more sophisticated CAR T cell approaches are going to be required for prostate metastasis than for B cell malignancies. Two main challenges that need to be resolved are how to increase the migration and infiltration of CAR T cells into prostate cancer bone metastases and how to counteract the immunosuppressive microenvironment found in bone lesions. Inclusion of homing (chemokine) receptors in CAR T cells may improve their recruitment to bone metastases, as may antibody-based combination therapies to normalize the tumor vasculature. Optimal activation of CAR T cells through the introduction of multiple costimulatory domains would help to overcome inhibitory signals from the tumor microenvironment. Likewise, combination therapy with checkpoint inhibitors that can reduce tumor immunosuppression may help improve efficacy. Other elegant approaches such as induced expression of immune stimulatory cytokines upon target recognition may also help to recruit other effector immune cells to metastatic sites. Although toxicities are difficult to predict in prostate cancer, severe on-target/off-tumor toxicities have been observed in clinical trials with use of CAR T cells against hematological malignancies; therefore, the choice of the target antigen is going to be crucial. This review focuses on different means of accomplishing maximal effectiveness of CAR T cell therapy for prostate cancer bone metastases while minimizing side effects and CAR T cell-associated toxicities. CAR T cell-based therapies for prostate cancer have the potential to be a therapy model for other solid tumors.
Collapse
|
664
|
Delivery of siRNA Using CXCR4-targeted Nanoparticles Modulates Tumor Microenvironment and Achieves a Potent Antitumor Response in Liver Cancer. Mol Ther 2015; 23:1772-1782. [PMID: 26278330 DOI: 10.1038/mt.2015.147] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 07/28/2015] [Indexed: 02/07/2023] Open
Abstract
Antiangiogenic therapy has recently emerged as a highly promising therapeutic strategy for treating hepatocellular carcinoma (HCC). However, the only clinically approved systemic antiangiogenic agent for advanced HCC is sorafenib, which exerts considerable toxicity. Moreover, acquired resistance to antiangiogenic therapy often develops and restricts the therapeutic efficacy of this treatment. Hence, in this study, we develop a CXCR4-targeted lipid-based nanoparticle (NP) formulation to specifically deliver vascular endothelial growth factor (VEGF) siRNA as an antiangiogenic substance into HCC. AMD3100, a CXCR4 antagonist, is added into NPs to serve as both a targeting moiety and a sensitizer to antiangiogenic therapy. We demonstrate that AMD-modified NPs (AMD-NPs) can efficiently deliver VEGF siRNAs into HCC and downregulate VEGF expression in vitro and in vivo. Despite the upregulation of the SDF1α/CXCR4 axis upon the induction of hypoxia after antiangiogenic therapy, CXCR4 inhibition by AMD-NPs in combination with either conventional sorafenib treatment or VEGF siRNA prevents the infiltration of tumor-associated macrophages. These dual treatments also induce synergistic antiangiogenic effects and suppress local and distant tumor growth in HCC. In conclusion, the tumor-targeted multifunctional AMD-NPs that co-deliver VEGF siRNA and AMD3100 provide an effective approach for overcoming tumor evasion of antiangiogenic therapy, leading to delayed tumor progression in HCC.
Collapse
|
665
|
Activation of neuroimmune pathways increases therapeutic effects of radiotherapy on poorly differentiated breast carcinoma. Brain Behav Immun 2015; 48:174-85. [PMID: 25736062 DOI: 10.1016/j.bbi.2015.02.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 02/13/2015] [Accepted: 02/23/2015] [Indexed: 12/11/2022] Open
Abstract
Recent studies document the importance of neuronal dysfunction in cancer development and metastasis. We reported previously that both depletion of neuropeptides in capsaicin-sensitive sensory nerve endings and vagotomy increases metastasis of triple negative breast carcinoma. Of the sensory neuropeptides, Substance P (SP) is distributed widely for regulation of immune functions. We therefore examined the affects of continuous exposure to low doses of SP on brain metastatic cells of the mouse breast carcinoma (4TBM) in the presence of radiotherapy (RT) thought to increase antigenicity of cancer cells. 4TBM cells have a cancer stem cell phenotype and induce extensive visceral metastasis after orthotopic inoculation into the mammary pad. Results demonstrated that SP treatment decreases the number of tumor-infiltrating myeloid-derived suppressor cells as well as the TNF-α response to LPS challenge. SP also increased CD4+Cd25(bright) cells in draining lymph nodes of tumor-bearing animals and IFN-γ secretion from leukocyte culture prepared from lymph nodes and spleens of tumor-bearing animals. SP also prevented tumor-induced degeneration of sensory nerve endings and altered release of angiogenic factors from cancer-associated fibroblasts (CAF) and tumor explants. In accordance with these observed immunological effects, combination treatment of continuous SP with a single dose of RT induced complete tumor regression and significantly reduced or prevented metastasis in 50% of the animals while suppressing primary tumor growth and metastasis in the remaining mice. These original findings demonstrate that SP through neuroimmune modulation can prevent formation of immune suppression in the tumor microenvironment, enhance cytotoxic immunity in the presence of RT and prevent metastatic growth.
Collapse
|
666
|
Kent D. The stereotypical molecular cascade in neovascular age-related macular degeneration: the role of dynamic reciprocity. Eye (Lond) 2015; 29:1416-26. [PMID: 26228288 DOI: 10.1038/eye.2015.140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 06/22/2015] [Indexed: 12/15/2022] Open
Abstract
This review summarises our current understanding of the molecular basis of subretinal neovascularisation (SRNV) in age-related macular degeneration (AMD). The term neovascular AMD (NVAMD) is derived from the dominant early clinical features of haemorrhage, fluid, and lipid in the subretinal space (SRS) and the historical role of fluorescein angiography in detecting the presence of NV tissue. However, at the cellular level, SRNV resembles an aberrant but stereotypical tissue repair response that incorporates both an early inflammatory phase and a late fibrotic phase in addition to the neovascular (NV) component that dominates the early clinical presentation. This review will seek not only to highlight the important molecules involved in each of these components but to demonstrate that the development of SRNV has its origins in the earliest events in non-NV AMD pathogenesis. Current evidence suggests that this early-stage pathogenesis is characterised by complement-mediated immune dysregulation, leading to a state of chronic inflammation in the retinal pigment epithelium/Bruch's membrane/choriocapillaris complex. These initial events can be seamlessly and inextricably linked to late-stage development of SRNV in AMD by the process of dynamic reciprocity (DyR), the ongoing bidirectional communication between cells, and their surrounding matrix. Moreover, this correlation between disease onset and eventual outcome is reflected in the temporal and spatial correlation between chronic inflammation, NV, and fibrosis within the reparative microenvironment of the SRS. In summary, the downstream consequences of the earliest dysfunctional molecular events in AMD can result in the late-stage entity we recognize clinically as SRNV and is characterized by a spectrum of predictable, related, and stereotypical processes referred to as DyR.
Collapse
Affiliation(s)
- D Kent
- The Vision Clinic, Kilkenny, Ireland.,Faculty of Health and Life Sciences, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| |
Collapse
|
667
|
Lemon CM, Karnas E, Han X, Bruns OT, Kempa TJ, Fukumura D, Bawendi MG, Jain RK, Duda DG, Nocera DG. Micelle-Encapsulated Quantum Dot-Porphyrin Assemblies as in Vivo Two-Photon Oxygen Sensors. J Am Chem Soc 2015; 137:9832-42. [PMID: 26149349 DOI: 10.1021/jacs.5b04765] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Micelles have been employed to encapsulate the supramolecular assembly of quantum dots with palladium(II) porphyrins for the quantification of O2 levels in aqueous media and in vivo. Förster resonance energy transfer from the quantum dot (QD) to the palladium porphyrin provides a means for signal transduction under both one- and two-photon excitation. The palladium porphyrins are sensitive to O2 concentrations in the range of 0-160 Torr. The micelle-encapsulated QD-porphyrin assemblies have been employed for in vivo multiphoton imaging and lifetime-based oxygen measurements in mice with chronic dorsal skinfold chambers or cranial windows. Our results establish the utility of the QD-micelle approach for in vivo biological sensing applications.
Collapse
Affiliation(s)
- Christopher M Lemon
- †Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, United States.,‡Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Elizabeth Karnas
- ‡Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Xiaoxing Han
- §Edwin L. Steele Laboratory for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, 100 Blossom Street, Cox-7, Boston, Massachusetts 02114, United States
| | - Oliver T Bruns
- ‡Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Thomas J Kempa
- †Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, United States
| | - Dai Fukumura
- §Edwin L. Steele Laboratory for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, 100 Blossom Street, Cox-7, Boston, Massachusetts 02114, United States
| | - Moungi G Bawendi
- ‡Department of Chemistry, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Rakesh K Jain
- §Edwin L. Steele Laboratory for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, 100 Blossom Street, Cox-7, Boston, Massachusetts 02114, United States
| | - Dan G Duda
- §Edwin L. Steele Laboratory for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, 100 Blossom Street, Cox-7, Boston, Massachusetts 02114, United States
| | - Daniel G Nocera
- †Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, United States
| |
Collapse
|
668
|
Sanlorenzo M, Vujic I, Moy A, Quaglino P, Fierro MT, Gammaitoni L, Carnevale-Schianca F, Aglietta M, Sangiolo D. Synergy of molecular targeted approaches and immunotherapy in melanoma: preclinical basis and clinical perspectives. Expert Opin Biol Ther 2015. [DOI: 10.1517/14712598.2015.1069272] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
669
|
Nowak-Sliwinska P, Clavel CM, Păunescu E, te Winkel MT, Griffioen AW, Dyson PJ. Antiangiogenic and Anticancer Properties of Bifunctional Ruthenium(II)–p-Cymene Complexes: Influence of Pendant Perfluorous Chains. Mol Pharm 2015; 12:3089-96. [DOI: 10.1021/acs.molpharmaceut.5b00417] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Patrycja Nowak-Sliwinska
- Angiogenesis
Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
- Institute
of Chemical Sciences and Engineering, Swiss Federal Institute of Technology (EPFL), CH-1015, Lausanne, Switzerland
| | - Catherine M. Clavel
- Institute
of Chemical Sciences and Engineering, Swiss Federal Institute of Technology (EPFL), CH-1015, Lausanne, Switzerland
| | - Emilia Păunescu
- Institute
of Chemical Sciences and Engineering, Swiss Federal Institute of Technology (EPFL), CH-1015, Lausanne, Switzerland
| | - Marije T. te Winkel
- Angiogenesis
Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
- Institute
of Chemical Sciences and Engineering, Swiss Federal Institute of Technology (EPFL), CH-1015, Lausanne, Switzerland
| | - Arjan W. Griffioen
- Angiogenesis
Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Paul J. Dyson
- Institute
of Chemical Sciences and Engineering, Swiss Federal Institute of Technology (EPFL), CH-1015, Lausanne, Switzerland
| |
Collapse
|
670
|
Molecular classification of gastric adenocarcinoma: translating new insights from the cancer genome atlas research network. Curr Treat Options Oncol 2015; 16:17. [PMID: 25813036 DOI: 10.1007/s11864-015-0331-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gastric cancer is a heterogenous cancer, which may be classified into several distinct subtypes based on pathology and epidemiology, each with different initiating pathological processes and each possibly having different tumor biology. A classification of gastric cancer should be important to select patients who can benefit from the targeted therapies or to precisely predict prognosis. The Cancer Genome Atlas (TCGA) study collaborated with previous reports regarding subtyping gastric cancer but also proposed a refined classification based on molecular characteristics. The addition of the new molecular classification strategy to a current classical subtyping may be a promising option, particularly stratification by Epstein-Barr virus (EBV) and microsatellite instability (MSI) statuses. According to TCGA study, EBV gastric cancer patients may benefit the programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) antibodies or phosphoinositide 3-kinase (PI3K) inhibitors which are now being developed. The discoveries of predictive biomarkers should improve patient care and individualized medicine in the management since the targeted therapies may have the potential to change the landscape of gastric cancer treatment, moreover leading to both better understanding of the heterogeneity and better outcomes. Patient enrichment by predictive biomarkers for new treatment strategies will be critical to improve clinical outcomes. Additionally, liquid biopsies will be able to enable us to monitor in real-time molecular escape mechanism, resulting in better treatment strategies.
Collapse
|
671
|
Deguchi A, Tomita T, Ohto U, Takemura K, Kitao A, Akashi-Takamura S, Miyake K, Maru Y. Eritoran inhibits S100A8-mediated TLR4/MD-2 activation and tumor growth by changing the immune microenvironment. Oncogene 2015; 35:1445-56. [DOI: 10.1038/onc.2015.211] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 04/30/2015] [Accepted: 05/04/2015] [Indexed: 01/08/2023]
|
672
|
Graybill W, Sood AK, Monk BJ, Coleman RL. State of the science: Emerging therapeutic strategies for targeting angiogenesis in ovarian cancer. Gynecol Oncol 2015; 138:223-6. [PMID: 26166806 DOI: 10.1016/j.ygyno.2015.07.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Whitney Graybill
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Medical University of South Carolina, Charleston, SC, United States
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Bradley J Monk
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Creighton University, Phoenix, AZ, United States
| | - Robert L Coleman
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.
| |
Collapse
|
673
|
|
674
|
Porcellini S, Asperti C, Valentinis B, Tiziano E, Mangia P, Bordignon C, Rizzardi GP, Traversari C. The tumor vessel targeting agent NGR-TNF controls the different stages of the tumorigenic process in transgenic mice by distinct mechanisms. Oncoimmunology 2015; 4:e1041700. [PMID: 26451306 DOI: 10.1080/2162402x.2015.1041700] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 04/10/2015] [Accepted: 04/10/2015] [Indexed: 10/23/2022] Open
Abstract
NGR-TNF is a vascular targeting agent in advanced clinical development, coupling tumor necrosis factor-α (TNF) with the CNGRCG peptide, which targets a CD13 isoform specifically expressed by angiogenic vessels. Antitumor efficacy of NGR-TNF has been described in different transplantation tumor models. Nevertheless, the mechanism underlying its activity is not fully understood. In the wild type and in the immunodeficient (RAG-/-) RIP1-Tag2 models of multistage pancreatic carcinogenesis, we demonstrate that CD13 is highly expressed on endothelial cells of hyperplastic and angiogenic islets, whereas its expression is down regulated in tumors where it partially colocalize with pericytes. In vivo CNGRCG peptides coupled to fluorescent nanoparticles (quantum dots) bind to CD13 and colocalize with anti-CD31, in pancreatic islets. At early stage, low doses of NGR-murine (m)TNF have a direct cytotoxic effect inducing endothelial cell apoptosis, reducing vessel density and eventually inhibiting the development of angiogenic islets. At a later stage, NGR-mTNF is able to reduce tumor growth inducing vascular normalization, exclusively when treatment is carried out in the immunocompetent mice. Interestingly, NGR-mTNF-treated tumors from these mice are characterized by CD8+ T cell infiltration. At molecular level, overexpression of genes involved in vessels normalization was detected only in NGR-mTNF-treated tumors from immunocompetent mice. These findings identified a new mechanism of action of NGR-mTNF, providing support for the development of new therapeutic strategies combining chemotherapy or active/adoptive immunotherapies to low dose NGR-TNF treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Claudio Bordignon
- MolMed SpA ; Milan, Italy ; Vita-Salute San Raffaele University ; Milan, Italy
| | | | | |
Collapse
|
675
|
Malvicini M, Fiore E, Ghiaccio V, Piccioni F, Rizzo M, Olmedo Bonadeo L, García M, Rodríguez M, Bayo J, Peixoto E, Atorrasagasti C, Alaniz L, Aquino J, Matar P, Mazzolini G. Tumor Microenvironment Remodeling by 4-Methylumbelliferone Boosts the Antitumor Effect of Combined Immunotherapy in Murine Colorectal Carcinoma. Mol Ther 2015; 23:1444-55. [PMID: 26105158 DOI: 10.1038/mt.2015.112] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 06/08/2015] [Indexed: 01/19/2023] Open
Abstract
We have previously demonstrated that a low dose of cyclophosphamide (Cy) combined with gene therapy of interleukin-12 (AdIL-12) has a synergistic, although limited, antitumoral effect in mice with colorectal carcinoma. The main mechanism involved in the efficacy of Cy+AdIL-12 was the induction of a specific immune response mediated by cytotoxic T lymphocytes. Our current aims were to evaluate the effects of 4-methylumbelliferone (4Mu), a selective inhibitor of hyaluronan (HA) synthesis, on tumor microenvironment (TME) and to investigate how 4Mu affects the therapeutic efficacy of Cy+AdIL-12. The results showed that 4Mu significantly reduced the amount of tumoral HA leading to a significant decrease in tumor interstitial pressure (TIP). As a consequence, tumor perfusion was improved allowing an increased adenoviral transgene expression. In addition, treatment with 4Mu boosted the number of cytotoxic T lymphocytes that reach the tumor after adoptive transfer resulting in a potent inhibition of tumor growth. Importantly, we observed complete tumor regression in 75% of mice when 4Mu was administrated in combination with Cy+AdIL-12. The triple combination 4Mu+Cy+AdIL-12 also induced a shift toward antiangiogenic factors production in tumor milieu. Our results showed that TME remodeling is an interesting strategy to increase the efficacy of anticancer immunotherapies based on gene and/or cell therapy.
Collapse
Affiliation(s)
- Mariana Malvicini
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina.,CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - Esteban Fiore
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina
| | - Valentina Ghiaccio
- Dipartamento di Sanità Pubblica, Medicina Clinica e Moleculare, Università degli studi di Cagliari, Sardegna, Italia
| | - Flavia Piccioni
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina
| | - Miguel Rizzo
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina
| | - Lucila Olmedo Bonadeo
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina
| | - Mariana García
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina.,CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - Marcelo Rodríguez
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina
| | - Juan Bayo
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina
| | - Estanislao Peixoto
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina
| | - Catalina Atorrasagasti
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina.,CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - Laura Alaniz
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina.,CIT NOBA, Universidad Nacional del Noroeste, Junín, Buenos Aires, Argentina
| | - Jorge Aquino
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina.,CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - Pablo Matar
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina.,Institute of Experimental Genetics, School of Medical Sciences, Universidad Nacional de Rosario, Rosario, Argentina
| | - Guillermo Mazzolini
- Gene Therapy Laboratory, School of Medicine, Universidad Austral, Buenos Aires, Argentina.,CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| |
Collapse
|
676
|
Huang D, Lan H, Liu F, Wang S, Chen X, Jin K, Mou X. Anti-angiogenesis or pro-angiogenesis for cancer treatment: focus on drug distribution. Int J Clin Exp Med 2015; 8:8369-8376. [PMID: 26309490 PMCID: PMC4537982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 06/07/2015] [Indexed: 06/04/2023]
Abstract
Enhancing chemotherapy delivery to tumors, improving tumor growth control, reducing metastasis, and increasing survival are all critical objectives of improved cancer therapy. One of the obstacles to the success of anticancer therapies is related to the inefficient distribution of drugs to tumor cells. To be effective, chemotherapeutics must reach a concentration in cancer cells that is sufficient to inhibit its targets. In the past years, the vascular normalization theory has gained widespread acceptance for explaining additional antitumor effects of inhibitors of vascular endothelial growth factor (VEGF) signaling, when combined with chemotherapeutics. Vascular normalization is a strategy to enhance the antitumor effects of chemotherapeutics, but this is time and dose dependent and therefore difficult to implement clinically. Thus, alternative strategies that overcome these issues are needed. Accumulating scientific data demonstrate an alternative approach called "vascular promotion therapy" can increase chemotherapeutics delivery and intracellular uptake of the drug and reduces hypoxia by increasing tumor blood vessel density, blood flow, leakiness, and dilation, which leads to reduced cancer growth and metastasis. In this article, we first summarize the structural and functional abnormalities of the tumor microvasculature to highlight the importance of this phenomenon for chemotherapeutics distribution. Next, we summarize the limitations of anti-angiogenic strategy in cancer treatment, discuss some key prototypical underlying mechanisms of vascular normalization and initial clinical evidence of vascular promotion therapy, and speculate on the clinical potential of anticoagulation as a novel paradigm to improve cancer treatment.
Collapse
Affiliation(s)
- Dongsheng Huang
- Clinical Research Institute, Zhejiang Provincial People’s HospitalHangzhou 310014, China
| | - Huanrong Lan
- Department of Gastrointestinal Surgery, Shaoxing People’s Hospital, Shaoxing Hospital of Zhejiang UniversityShaoxing 312000, Zhejiang Province, P.R. China
| | - Fanlong Liu
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhou 310003, Zhejiang, P.R. China
| | - Shibing Wang
- Clinical Research Institute, Zhejiang Provincial People’s HospitalHangzhou 310014, China
| | - Xiaoyi Chen
- Clinical Research Institute, Zhejiang Provincial People’s HospitalHangzhou 310014, China
| | - Ketao Jin
- Clinical Research Institute, Zhejiang Provincial People’s HospitalHangzhou 310014, China
- Department of Gastrointestinal Surgery, Shaoxing People’s Hospital, Shaoxing Hospital of Zhejiang UniversityShaoxing 312000, Zhejiang Province, P.R. China
| | - Xiaozhou Mou
- Clinical Research Institute, Zhejiang Provincial People’s HospitalHangzhou 310014, China
| |
Collapse
|
677
|
Yuan J, Zhou J, Dong Z, Tandon S, Kuk D, Panageas KS, Wong P, Wu X, Naidoo J, Page DB, Wolchok JD, Hodi FS. Pretreatment serum VEGF is associated with clinical response and overall survival in advanced melanoma patients treated with ipilimumab. Cancer Immunol Res 2015; 2:127-32. [PMID: 24778276 DOI: 10.1158/2326-6066.cir-13-0163] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Ipilimumab, an antibody that blocks CTL antigen 4 (CTLA-4), improves overall survival (OS) for patients with metastatic melanoma. Given its role in angiogenesis and immune evasion, serum VEGF levels were evaluated for association with clinical benefit in ipilimumab-treated patients. Sera were collected from 176 patients treated at 3 (n = 98) or 10 mg/kg (n = 68). The VEGF levels before treatment and at induction completion (week 12) were analyzed using the Meso Scale Discovery kit. The association of the levels of VEGF with clinical responses and OS were assessed using the Fisher exact and Kaplan-Meier log-rank tests. VEGF as a continuous variable was associated with OS (P = 0.002). Using 43 pg/mL as the cutoff pretreatment VEGF value defined by maximally selected log-rank statistics, pretreatment VEGF values correlated with clinical benefit at week 24 (P = 0.019; 159 patients evaluable). Pretreatment VEGF ≥ 43 pg/mL was associated with decreased OS (median OS 6.6 vs. 12.9 months, P = 0.006; 7.4 vs. 14.3 months, P = 0.037 for 3 mg/kg; and 6.2 vs. 10.9 months, P = 0.048 for 10 mg/kg). There was no correlation between VEGF changes and clinical outcome. Serum VEGF may be a predictive biomarker for ipilimumab treatment and is worthy of prospective investigation with various forms of immunologic checkpoint blockade.
Collapse
Affiliation(s)
- Jianda Yuan
- Authors' Affiliations Melanoma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
678
|
Johansson A, Hamzah J, Ganss R. More than a scaffold: Stromal modulation of tumor immunity. Biochim Biophys Acta Rev Cancer 2015; 1865:3-13. [PMID: 26071879 DOI: 10.1016/j.bbcan.2015.06.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 05/20/2015] [Accepted: 06/04/2015] [Indexed: 12/25/2022]
Abstract
Current clinical success with anti-cancer immunotherapy provides exciting new treatment opportunities. While encouraging, more needs to be done to induce durable effects in a higher proportion of patients. Increasing anti-tumor effector T cell quantity or quality alone does not necessarily correlate with therapeutic outcome. Instead, the tumor microenvironment is a critical determinant of anti-cancer responsiveness to immunotherapy and can confer profound resistance. Yet, the tumor-promoting environment - due to its enormous plasticity - also delivers the best opportunities for adjuvant therapy aiming at recruiting, priming and sustaining anti-tumor cytotoxicity. While the tumor environment as an entity is increasingly well understood, current interventions are still broad and often systemic. In contrast, tumors grow in a highly compartmentalized environment which includes the vascular/perivascular niche, extracellular matrix components and in some tumors lymph node aggregates; all of these structures harbor and instruct subsets of immune cells. Targeting and re-programming specific compartments may provide better opportunities for adjuvant immunotherapy.
Collapse
Affiliation(s)
- Anna Johansson
- Vascular Biology and Stromal Targeting, Harry Perkins Institute of Medical Research, The University of Western Australia, Centre for Medical Research, Nedlands, Western Australia 6009, Australia
| | - Juliana Hamzah
- Targeted Drug Delivery, Imaging and Therapy, Harry Perkins Institute of Medical Research, The University of Western Australia, Centre for Medical Research, Nedlands, Western Australia 6009, Australia
| | - 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 6009, Australia.
| |
Collapse
|
679
|
Leblond MM, Gérault AN, Corroyer-Dulmont A, MacKenzie ET, Petit E, Bernaudin M, Valable S. Hypoxia induces macrophage polarization and re-education toward an M2 phenotype in U87 and U251 glioblastoma models. Oncoimmunology 2015; 5:e1056442. [PMID: 26942063 PMCID: PMC4760330 DOI: 10.1080/2162402x.2015.1056442] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/22/2015] [Accepted: 05/24/2015] [Indexed: 11/28/2022] Open
Abstract
Hypoxia is a common feature of solid tumors, particularly in glioblastoma (GBM), and known to be a poor prognosis factor in GBM patients. The growth of GBM is also associated with a marked inflammation partially characterized by an accumulation of macrophage (MΦ) of the M2 phenotype. However, the transition between M1 MΦ (antitumoral) and M2 MΦ (protumoral) phenotypes is a dynamic process. We made the assumption that oxygen (O2) availability could be a major regulator of this transition and that the intratumoral O2 gradient is of importance. We evaluated, in vivo, the impact of hypoxia on MΦ tropism and polarization in two models of human GBM, well differentiated by their degree of hypoxia. MΦ migration in the tumor was more pronounced in the more hypoxic tumor of the two GBM models. In the more hypoxic of the models, we have shown that MΦ migrated at the tumor site only when hypoxia takes place. We also demonstrated that the acquisition of the M2 phenotype was clearly an evolving phenomenon with hypoxia as the major trigger for this transition. In support of these in vivo finding, M0 but also M1 MΦ cultured in moderate or severe hypoxia displayed a phenotype close to that of M2 MΦ whose phenotype was further reinforced by severe hypoxia. These results highlight the role of hypoxia in the aggressiveness of GBM, in part, by transforming MΦ such that a protumoral activity is expressed.
Collapse
Affiliation(s)
- Marine M Leblond
- CNRS; UMR6301-ISTCT; CERVOxy Group; GIP CYCERON; Bd Henri Becquerel; Caen Cedex, France
- Université de Caen Basse-Normandie; Caen, France
- CEA; DSV/I2BM; Caen, France
- Normandie Université; Caen, France
| | - Aurélie N Gérault
- CNRS; UMR6301-ISTCT; CERVOxy Group; GIP CYCERON; Bd Henri Becquerel; Caen Cedex, France
- Université de Caen Basse-Normandie; Caen, France
- CEA; DSV/I2BM; Caen, France
- Normandie Université; Caen, France
| | - Aurélien Corroyer-Dulmont
- CNRS; UMR6301-ISTCT; CERVOxy Group; GIP CYCERON; Bd Henri Becquerel; Caen Cedex, France
- Université de Caen Basse-Normandie; Caen, France
- CEA; DSV/I2BM; Caen, France
- Normandie Université; Caen, France
| | - Eric T MacKenzie
- CNRS; UMR6301-ISTCT; CERVOxy Group; GIP CYCERON; Bd Henri Becquerel; Caen Cedex, France
- Université de Caen Basse-Normandie; Caen, France
- CEA; DSV/I2BM; Caen, France
- Normandie Université; Caen, France
| | - Edwige Petit
- CNRS; UMR6301-ISTCT; CERVOxy Group; GIP CYCERON; Bd Henri Becquerel; Caen Cedex, France
- Université de Caen Basse-Normandie; Caen, France
- CEA; DSV/I2BM; Caen, France
- Normandie Université; Caen, France
| | - Myriam Bernaudin
- CNRS; UMR6301-ISTCT; CERVOxy Group; GIP CYCERON; Bd Henri Becquerel; Caen Cedex, France
- Université de Caen Basse-Normandie; Caen, France
- CEA; DSV/I2BM; Caen, France
- Normandie Université; Caen, France
| | - Samuel Valable
- CNRS; UMR6301-ISTCT; CERVOxy Group; GIP CYCERON; Bd Henri Becquerel; Caen Cedex, France
- Université de Caen Basse-Normandie; Caen, France
- CEA; DSV/I2BM; Caen, France
- Normandie Université; Caen, France
| |
Collapse
|
680
|
Peske JD, Woods AB, Engelhard VH. Control of CD8 T-Cell Infiltration into Tumors by Vasculature and Microenvironment. Adv Cancer Res 2015. [PMID: 26216636 DOI: 10.1016/bs.acr.2015.05.001] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
CD8 T-cells are a critical brake on the initial development of tumors. In established tumors, the presence of CD8 T-cells is correlated with a positive patient prognosis, although immunosuppressive mechanisms limit their effectiveness and they are rarely curative without manipulation. Cancer immunotherapies aim to shift the balance back to dominant antitumor immunity through antibody blockade of immunosuppressive signaling pathways, vaccination, and adoptive transfer of activated or engineered T-cells. These approaches have yielded striking responses in small subsets of patients with solid tumors, most notably those with melanoma. Importantly, the subset of patients who respond to vaccination or immunosuppression blockade therapies are those with CD8 T-cells present in the tumor prior to initiating therapy. While current adoptive cell therapy approaches can be dramatically effective, they require infusion of extremely large numbers of T-cells, but the number that actually infiltrates the tumor is very small. Thus, poor representation of CD8 T-cells in tumors is a fundamental hurdle to successful immunotherapy, over and above the well-established barrier of immunosuppression. In this review, we discuss the factors that determine whether immune cells are present in tumors, with a focus on the representation of cytotoxic CD8 T-cells. We emphasize the critically important role of tumor-associated vasculature as a gateway that enables the active infiltration of both effector and naïve CD8 T-cells that exert antitumor activity. We also discuss strategies to enhance the gateway function and extend the effectiveness of immunotherapies to a broader set of cancer patients.
Collapse
Affiliation(s)
- J David Peske
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Amber B Woods
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Victor H Engelhard
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA.
| |
Collapse
|
681
|
Chen Y, Duda DG. Targeting immunosuppression after standard sorafenib treatment to facilitate immune checkpoint blockade in hepatocellular carcinoma - an auto-commentary on clinical potential and future development. Oncoimmunology 2015; 4:e1029703. [PMID: 26451297 DOI: 10.1080/2162402x.2015.1029703] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 03/07/2015] [Accepted: 03/11/2015] [Indexed: 01/21/2023] Open
Abstract
Immunotherapy has shown great promise to transform solid cancer treatment. The challenge is to optimally incorporate novel immunotherapeutics, such as immune checkpoint blockers, with standard therapies. This is well exemplified by multimodal therapies recently developed for liver cancer in which immunomodulation using CXCR4 inhibition prevented immunosuppression and enhanced sorafenib and anti-PD-1 therapeutic outcome.
Collapse
Affiliation(s)
- Yunching Chen
- Institute of Biomedical Engineering; National Tsing Hua University ; Hsinchu, Taiwan ; Steele Laboratories for Tumor Biology; Department of Radiation Oncology; Massachusetts General Hospital and Harvard Medical School ; Boston, MA USA
| | - Dan G Duda
- Steele Laboratories for Tumor Biology; Department of Radiation Oncology; Massachusetts General Hospital and Harvard Medical School ; Boston, MA USA
| |
Collapse
|
682
|
Suarez-Carmona M, Bourcy M, Lesage J, Leroi N, Syne L, Blacher S, Hubert P, Erpicum C, Foidart JM, Delvenne P, Birembaut P, Noël A, Polette M, Gilles C. Soluble factors regulated by epithelial-mesenchymal transition mediate tumour angiogenesis and myeloid cell recruitment. J Pathol 2015; 236:491-504. [PMID: 25880038 DOI: 10.1002/path.4546] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 03/18/2015] [Accepted: 04/13/2015] [Indexed: 01/13/2023]
Abstract
Epithelial-mesenchymal transition (EMT) programmes provide cancer cells with invasive and survival capacities that might favour metastatic dissemination. Whilst signalling cascades triggering EMT have been extensively studied, the impact of EMT on the crosstalk between tumour cells and the tumour microenvironment remains elusive. We aimed to identify EMT-regulated soluble factors that facilitate the recruitment of host cells in the tumour. Our findings indicate that EMT phenotypes relate to the induction of a panel of secreted mediators, namely IL-8, IL-6, sICAM-1, PAI-1 and GM-CSF, and implicate the EMT-transcription factor Snail as a regulator of this process. We further show that EMT-derived soluble factors are pro-angiogenic in vivo (in the mouse ear sponge assay), ex vivo (in the rat aortic ring assay) and in vitro (in a chemotaxis assay). Additionally, conditioned medium from EMT-positive cells stimulates the recruitment of myeloid cells. In a bank of 40 triple-negative breast cancers, tumours presenting features of EMT were significantly more angiogenic and infiltrated by a higher quantity of myeloid cells compared to tumours with little or no EMT. Taken together, our results show that EMT programmes trigger the expression of soluble mediators in cancer cells that stimulate angiogenesis and recruit myeloid cells in vivo, which might in turn favour cancer spread.
Collapse
Affiliation(s)
- Meggy Suarez-Carmona
- Laboratory of Tumour and Development Biology (LBTD), GIGA-Cancer, Liège, Belgium.,Laboratory of Experimental Pathology (LEP), GIGA-Cancer, Liège, Belgium
| | - Morgane Bourcy
- Laboratory of Tumour and Development Biology (LBTD), GIGA-Cancer, Liège, Belgium
| | - Julien Lesage
- INSERM UMR-S 903, Laboratoire Pol Bouin, University of Reims, France
| | - Natacha Leroi
- Laboratory of Tumour and Development Biology (LBTD), GIGA-Cancer, Liège, Belgium
| | - Laïdya Syne
- Laboratory of Tumour and Development Biology (LBTD), GIGA-Cancer, Liège, Belgium
| | - Silvia Blacher
- Laboratory of Tumour and Development Biology (LBTD), GIGA-Cancer, Liège, Belgium
| | - Pascale Hubert
- Laboratory of Experimental Pathology (LEP), GIGA-Cancer, Liège, Belgium
| | - Charlotte Erpicum
- Laboratory of Experimental Pathology (LEP), GIGA-Cancer, Liège, Belgium
| | - Jean-Michel Foidart
- Laboratory of Tumour and Development Biology (LBTD), GIGA-Cancer, Liège, Belgium
| | - Philippe Delvenne
- Laboratory of Experimental Pathology (LEP), GIGA-Cancer, Liège, Belgium
| | | | - Agnès Noël
- Laboratory of Tumour and Development Biology (LBTD), GIGA-Cancer, Liège, Belgium
| | - Myriam Polette
- INSERM UMR-S 903, Laboratoire Pol Bouin, University of Reims, France
| | - Christine Gilles
- Laboratory of Tumour and Development Biology (LBTD), GIGA-Cancer, Liège, Belgium
| |
Collapse
|
683
|
AECHL-1, a novel triterpenoid, targets tumor neo-vasculature and impairs the endothelial cell cytoskeleton. Angiogenesis 2015; 18:283-99. [PMID: 25952529 PMCID: PMC4472952 DOI: 10.1007/s10456-015-9466-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 04/29/2015] [Indexed: 11/26/2022]
Abstract
Tumor angiogenesis is characterized by abnormal vessel morphology leading to erratic and insufficient delivery of chemotherapeutics and oxygen, making the tumor core not only highly hypoxic but also unresponsive toward treatment. Such hypoxic conditions promote tumor aggressiveness, leading to the establishment of metastatic disease. Most anti-angiogenic treatments aim toward the destruction of tumor vasculature, which proves countereffective by further increasing its aggressive nature. Hence, developing drugs which target or regulate these processes might lead to a better delivery of chemotherapeutics resulting in tumor shrinkage. Plant-derived natural compounds having a bioactive ingredient, especially triterpenoids, have been known to possess anticancer properties. AECHL-1, a recently isolated novel triterpenoid with proven anticancer potential, is seemingly noncytotoxic toward HEK 293 and HUVECs. Also, cytotoxicity was absent during in vivo studies involving intraperitoneal injections with 5 µg/kg body weight AECHL-1 on SCID mice. When used at subtoxic doses, it was found to be effective in suppression of neo-vessel formation as demonstrated in the chick chorioallantoic membrane, rat aortic rings, Matrigel plugs and xenograft tumors implanted in SCID mice. Tumor vasculature from AECHL-1-treated mice showed greater mural cell coverage and relatively normalized architecture. Investigations into the molecular mechanisms responsible for these observations revealed an effect on the actin cytoskeleton of stimulated HUVECs as well as the VEGFR2-mediated MAPK pathway. AECHL-1 could effectively distinguish between stimulated and nonstimulated endothelial cells. AECHL-1 could also downregulate HIF-1α expression and VEGF secretion under hypoxic conditions, thus reducing the fears of unnecessarily aggravating tumor metastasis as a result of anti-angiogenic therapy. Results obtained from the aforementioned studies make it clear that though AECHL-1 shows promise in discouraging and pruning neo-vasculature, it may not affect existing vasculature as the doses used for the assays are significantly lower than the ones causing endothelial cell death and has potential to be considered as a candidate for therapeutic drug development.
Collapse
|
684
|
Ager A, May MJ. Understanding high endothelial venules: Lessons for cancer immunology. Oncoimmunology 2015; 4:e1008791. [PMID: 26155419 PMCID: PMC4485764 DOI: 10.1080/2162402x.2015.1008791] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 01/09/2015] [Accepted: 01/10/2015] [Indexed: 01/06/2023] Open
Abstract
High endothelial venules (HEVs) are blood vessels especially adapted for lymphocyte trafficking which are normally found in secondary lymphoid organs such as lymph nodes (LN) and Peyer's patches. It has long been known that HEVs develop in non-lymphoid organs during chronic inflammation driven by autoimmunity, infection or allografts. More recently, HEVs have been observed in solid, vascularized tumors and their presence correlated with reduced tumor size and improved patient outcome. It is proposed that newly formed HEV promote antitumor immunity by recruiting naive lymphocytes into the tumor, thus allowing the local generation of cancerous tissue-destroying lymphocytes. Understanding how HEVs develop and function are therefore important to unravel their role in human cancers. In LN, HEVs develop during embryonic and early post-natal life and are actively maintained by the LN microenvironment. Systemic blockade of lymphotoxin-β receptor leads to HEV de-differentiation, but the LN components that induce HEV differentiation have remained elusive. Recent elegant studies using gene-targeted mice have demonstrated clearly that triggering the lymphotoxin-β receptor in endothelial cells (EC) induces the differentiation of HEV and that CD11c+ dendritic cells play a crucial role in this process. It will be important to determine whether lymphotoxin-β receptor-dependent signaling in EC drives the development of HEV during tumorigenesis and which cells have HEV-inducer properties. This may reveal therapeutic approaches to promote HEV neogenesis and determine the impact of newly formed HEV on tumor immunity.
Collapse
Key Words
- EC, endothelial cells
- FRC, fibroblast reticular cells
- HEC, high endothelial cells
- HEV, high endothelial venules
- LN, lymph nodes
- LPA, lysophosphatidic acid
- LT, lymphotoxin
- LT-βR, lymphotoxin-β receptor
- MAdCAM, mucosal cell adhesion molecule
- PNAd, peripheral node addressin
- SIP, sphingosine-1-phosphate
- T cell homing
- TLO, tertiary lymphoid organ
- VE-cadherin, vascular endothelial cadherin
- VEGF, vascular endothelial growth factor
- dendritic cells
- high endothelial venules
- lymphotoxin-β receptor
- tumor immunotherapy
Collapse
Affiliation(s)
- Ann Ager
- Infection and Immunity; School of Medicine; Cardiff University ; Cardiff, UK
| | - Michael J May
- School of Veterinary Medicine; University of Pennsylvania ; Philadelphia, PA, USA
| |
Collapse
|
685
|
Chen Y, Ramjiawan RR, Reiberger T, Ng MR, Hato T, Huang Y, Ochiai H, Kitahara S, Unan EC, Reddy TP, Fan C, Huang P, Bardeesy N, Zhu AX, Jain RK, Duda DG. CXCR4 inhibition in tumor microenvironment facilitates anti-programmed death receptor-1 immunotherapy in sorafenib-treated hepatocellular carcinoma in mice. Hepatology 2015; 61:1591-602. [PMID: 25529917 PMCID: PMC4406806 DOI: 10.1002/hep.27665] [Citation(s) in RCA: 338] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 12/16/2014] [Indexed: 12/15/2022]
Abstract
UNLABELLED Sorafenib, a broad tyrosine kinase inhibitor, is the only approved systemic therapy for advanced hepatocellular carcinoma (HCC) but provides limited survival benefits. Recently, immunotherapy has emerged as a promising treatment strategy, but its role remains unclear in HCCs, which are associated with decreased cytotoxic CD8(+) T-lymphocyte infiltration in both murine and human tumors. Moreover, in mouse models after sorafenib treatment intratumoral hypoxia is increased and may fuel evasive resistance. Using orthotopic HCC models, we now show that increased hypoxia after sorafenib treatment promotes immunosuppression, characterized by increased intratumoral expression of the immune checkpoint inhibitor programmed death ligand-1 and accumulation of T-regulatory cells and M2-type macrophages. We also show that the recruitment of immunosuppressive cells is mediated in part by hypoxia-induced up-regulation of stromal cell-derived 1 alpha. Inhibition of the stromal cell-derived 1 alpha receptor (C-X-C receptor type 4 or CXCR4) using AMD3100 prevented the polarization toward an immunosuppressive microenvironment after sorafenib treatment, inhibited tumor growth, reduced lung metastasis, and improved survival. However, the combination of AMD3100 and sorafenib did not significantly change cytotoxic CD8(+) T-lymphocyte infiltration into HCC tumors and did not modify their activation status. In separate experiments, antibody blockade of the programmed death ligand-1 receptor programmed death receptor-1 (PD-1) showed antitumor effects in treatment-naive tumors in orthotopic (grafted and genetically engineered) models of HCC. However, anti-PD-1 antibody treatment had additional antitumor activity only when combined with sorafenib and AMD3100 and not when combined with sorafenib alone. CONCLUSION Anti-PD-1 treatment can boost antitumor immune responses in HCC models; when used in combination with sorafenib, anti-PD-1 immunotherapy shows efficacy only with concomitant targeting of the hypoxic and immunosuppressive microenvironment with agents such as CXCR4 inhibitors.
Collapse
Affiliation(s)
- Yunching Chen
- E.L Steele Laboratory for Tumor Biology, Dept. of Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA,Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Rakesh R. Ramjiawan
- E.L Steele Laboratory for Tumor Biology, Dept. of Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA,Angiogenesis Laboratory, Cancer Center Amsterdam, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Thomas Reiberger
- E.L Steele Laboratory for Tumor Biology, Dept. of Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA,Division of Gastroenterology & Hepatology, Medical University of Vienna, Vienna, Austria
| | - Mei R. Ng
- E.L Steele Laboratory for Tumor Biology, Dept. of Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - Tai Hato
- E.L Steele Laboratory for Tumor Biology, Dept. of Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - Yuhui Huang
- E.L Steele Laboratory for Tumor Biology, Dept. of Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - Hiroki Ochiai
- E.L Steele Laboratory for Tumor Biology, Dept. of Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - Shuji Kitahara
- E.L Steele Laboratory for Tumor Biology, Dept. of Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - Elizabeth C. Unan
- E.L Steele Laboratory for Tumor Biology, Dept. of Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - Tejaswini P. Reddy
- E.L Steele Laboratory for Tumor Biology, Dept. of Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - Christopher Fan
- E.L Steele Laboratory for Tumor Biology, Dept. of Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - Peigen Huang
- E.L Steele Laboratory for Tumor Biology, Dept. of Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - Nabeel Bardeesy
- Department of Medicine, Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Andrew X. Zhu
- Department of Medicine, Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Rakesh K. Jain
- E.L Steele Laboratory for Tumor Biology, Dept. of Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - Dan G. Duda
- E.L Steele Laboratory for Tumor Biology, Dept. of Radiation Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA,Corresponding author: Dan G. Duda, DMD, PhD, Steele Laboratory for Tumor Biology, Massachusetts General Hospital, Cox-734, 100 Blossom Street, Boston, MA 02114; phone: (617) 726-4648; fax: (617) 726-1962;
| |
Collapse
|
686
|
Abstract
Effective immunotherapy promotes the killing of cancer cells by cytotoxic T cells. This requires not only that cancer-specific T cells be generated, but also that these T cells physically contact cancer cells. The coexistence in some patients of cancer cells and T cells that recognize them indicates that tumors may exhibit the phenomenon of immune privilege, in which immunogenic tissue is protected from immune attack. Here, we review the evidence that stromal cells of the tumor microenvironment mediate this restriction by excluding T cells from the vicinity of cancer cells. Overcoming this T cell checkpoint may thus enable optimal immunotherapy.
Collapse
Affiliation(s)
- Johanna A Joyce
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Douglas T Fearon
- Cold Spring Harbor Laboratory, New York, NY 11724, USA. Department of Microbiology and Immunology, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical School, New York, NY 10065, USA.
| |
Collapse
|
687
|
Apetoh L, Ladoire S, Coukos G, Ghiringhelli F. Combining immunotherapy and anticancer agents: the right path to achieve cancer cure? Ann Oncol 2015; 26:1813-1823. [PMID: 25922066 DOI: 10.1093/annonc/mdv209] [Citation(s) in RCA: 195] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 04/17/2015] [Indexed: 12/31/2022] Open
Abstract
Recent clinical trials revealed the impressive efficacy of immunological checkpoint blockade in different types of metastatic cancers. Such data underscore that immunotherapy is one of the most promising strategies for cancer treatment. In addition, preclinical studies provide evidence that some cytotoxic drugs have the ability to stimulate the immune system, resulting in anti-tumor immune responses that contribute to clinical efficacy of these agents. These observations raise the hypothesis that the next step for cancer treatment is the combination of cytotoxic agents and immunotherapies. The present review aims to summarize the immune-mediated effects of chemotherapeutic agents and their clinical relevance, the biological and clinical features of immune checkpoint blockers and finally, the preclinical and clinical rationale for novel therapeutic strategies combining anticancer agents and immune checkpoint blockers.
Collapse
Affiliation(s)
- L Apetoh
- Lipids, Nutrition, Cancer, INSERM, U866, Dijon; Department of Medicine, Université de Bourgogne, Dijon; Department of Oncology, Centre Georges François Leclerc, Dijon, France
| | - S Ladoire
- Lipids, Nutrition, Cancer, INSERM, U866, Dijon; Department of Medicine, Université de Bourgogne, Dijon; Department of Oncology, Centre Georges François Leclerc, Dijon, France
| | - G Coukos
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - F Ghiringhelli
- Lipids, Nutrition, Cancer, INSERM, U866, Dijon; Department of Medicine, Université de Bourgogne, Dijon; Department of Oncology, Centre Georges François Leclerc, Dijon, France.
| |
Collapse
|
688
|
Eosinophils orchestrate cancer rejection by normalizing tumor vessels and enhancing infiltration of CD8(+) T cells. Nat Immunol 2015; 16:609-17. [PMID: 25915731 DOI: 10.1038/ni.3159] [Citation(s) in RCA: 358] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 03/26/2015] [Indexed: 02/07/2023]
Abstract
Tumor-associated eosinophilia is frequently observed in cancer. However, despite numerous studies of patients with cancer and mouse models of cancer, it has remained uncertain if eosinophils contribute to tumor immunity or are mere bystander cells. Here we report that activated eosinophils were essential for tumor rejection in the presence of tumor-specific CD8(+) T cells. Tumor-homing eosinophils secreted chemoattractants that guided T cells into the tumor, which resulted in tumor eradication and survival. Activated eosinophils initiated substantial changes in the tumor microenvironment, including macrophage polarization and normalization of the tumor vasculature, which are known to promote tumor rejection. Thus, our study presents a new concept for eosinophils in cancer that may lead to novel therapeutic strategies.
Collapse
|
689
|
Tissue resident regulatory T cells: novel therapeutic targets for human disease. Cell Mol Immunol 2015; 12:543-52. [PMID: 25891216 PMCID: PMC4579654 DOI: 10.1038/cmi.2015.23] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Revised: 02/25/2015] [Accepted: 02/25/2015] [Indexed: 02/07/2023] Open
Abstract
Over the past decade, the ability of regulatory T cells (Tregs) to suppress multiple types of immune cells has received tremendous attention. Mounting evidence has revealed that tissue resident Tregs control non-immunological processes of their target tissues and contribute to a plethora of human diseases. The identification of novel tissue-specific Tregs has highlighted their heterogeneity and complexity. This review summarizes the recent findings for visceral adipose tissue CD4+Foxp3+ regulatory T cells (VAT Tregs), muscle Tregs, bone Tregs and skin memory Tregs, with a focus on their unique functions in local tissues. This interpretation of the roles of tissue-specific Tregs and of their involvement in disease progression provides new insight into the discovery of potential therapeutic targets of human diseases.
Collapse
|
690
|
Rivera LB, Meyronet D, Hervieu V, Frederick MJ, Bergsland E, Bergers G. Intratumoral myeloid cells regulate responsiveness and resistance to antiangiogenic therapy. Cell Rep 2015; 11:577-91. [PMID: 25892230 DOI: 10.1016/j.celrep.2015.03.055] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 01/02/2015] [Accepted: 03/25/2015] [Indexed: 12/21/2022] Open
Abstract
Antiangiogenic therapy is commonly used in the clinic, but its beneficial effects are short-lived, leading to tumor relapse within months. Here, we found that the efficacy of angiogenic inhibitors targeting the VEGF/VEGFR pathway was dependent on induction of the angiostatic and immune-stimulatory chemokine CXCL14 in mouse models of pancreatic neuroendocrine and mammary tumors. In response, tumors reinitiated angiogenesis and immune suppression by activating PI3K signaling in all CD11b+ cells, rendering tumors nonresponsive to VEGF/VEGFR inhibition. Adaptive resistance was also associated with an increase in Gr1+CD11b+ cells, but targeting Gr1+ cells was not sufficient to further sensitize angiogenic blockade because tumor-associated macrophages (TAMs) would compensate for the lack of such cells and vice versa, leading to an oscillating pattern of distinct immune-cell populations. However, PI3K inhibition in CD11b+ myeloid cells generated an enduring angiostatic and immune-stimulatory environment in which antiangiogenic therapy remained efficient.
Collapse
Affiliation(s)
- Lee B Rivera
- Department of Neurological Surgery, Brain Tumor Research Center, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - David Meyronet
- Université Lyon 1, Centre de Pathologie et Neuropathologie Est, Hospices Civils de Lyon, Bron Cedex 69677, France
| | - Valérie Hervieu
- Université Lyon 1, Service d'Anatomie Pathologique, Hôpital Édouard Herriot, Hospices Civils de Lyon, Lyon Cedex 69003, France
| | - Mitchell J Frederick
- Department of Head and Neck Surgery, Research Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Emily Bergsland
- Department of Medicine, UCSF Mount Zion Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Gabriele Bergers
- Department of Neurological Surgery, Brain Tumor Research Center, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
691
|
Nouraee N, Mowla SJ, Calin GA. Tracking miRNAs' footprints in tumor-microenvironment interactions: Insights and implications for targeted cancer therapy. Genes Chromosomes Cancer 2015; 54:335-52. [PMID: 25832733 DOI: 10.1002/gcc.22244] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 01/16/2015] [Accepted: 01/17/2015] [Indexed: 12/16/2022] Open
Abstract
In the past decades, cancer medicine studies have mainly focused on tumor cell biology as the main promoter of solid tumor progression. However, tumor biology does not explain the intertwinement and ambiguity of the tumors' territory. Recently, the approach of understanding cancer has shifted from investigating the biology of tumor cells to studying the microenvironment surrounding them. MicroRNAs (miRNAs), which play a role in exploiting indigenous stromal cells and are components that cooperate and produce a favorable microenvironment for progressive tumor formation, have been implicated in numerous processes essential for tumor initiation and growth. Understanding the mechanisms underlying interactions between tumor cells and their adjacent environment holds many promises for the future of cancer-targeted therapies. Herein, we provide a step-by-step account of miRNA involvement in tumor-microenvironment interactions as the micromediators of tumor cell and stroma communications. We also focus on the clinical challenges in using miRNAs tof overcome therapy resistance mechanisms and tumor heterogeneity bias in cancer therapy.
Collapse
Affiliation(s)
- Nazila Nouraee
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | | |
Collapse
|
692
|
Jain RK, Martin JD, Stylianopoulos T. The role of mechanical forces in tumor growth and therapy. Annu Rev Biomed Eng 2015; 16:321-46. [PMID: 25014786 DOI: 10.1146/annurev-bioeng-071813-105259] [Citation(s) in RCA: 603] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tumors generate physical forces during growth and progression. These physical forces are able to compress blood and lymphatic vessels, reducing perfusion rates and creating hypoxia. When exerted directly on cancer cells, they can increase cells' invasive and metastatic potential. Tumor vessels-while nourishing the tumor-are usually leaky and tortuous, which further decreases perfusion. Hypoperfusion and hypoxia contribute to immune evasion, promote malignant progression and metastasis, and reduce the efficacy of a number of therapies, including radiation. In parallel, vessel leakiness together with vessel compression causes a uniformly elevated interstitial fluid pressure that hinders delivery of blood-borne therapeutic agents, lowering the efficacy of chemo- and nanotherapies. In addition, shear stresses exerted by flowing blood and interstitial fluid modulate the behavior of cancer and a variety of host cells. Taming these physical forces can improve therapeutic outcomes in many cancers.
Collapse
Affiliation(s)
- Rakesh K Jain
- Edwin L. Steele Laboratory, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114;
| | | | | |
Collapse
|
693
|
Madorsky Rowdo FP, Baron A, Urrutia M, Mordoh J. Immunotherapy in Cancer: A Combat between Tumors and the Immune System; You Win Some, You Lose Some. Front Immunol 2015; 6:127. [PMID: 25859247 PMCID: PMC4374472 DOI: 10.3389/fimmu.2015.00127] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 03/08/2015] [Indexed: 12/18/2022] Open
Abstract
Cancer immunotherapy has emerged as a treatment modality, mainly as the result of discoveries in the immune response regulation, including mechanisms that turn off immune responses. Immunogenic cutaneous melanoma is a canonical model for therapeutic immunotherapy studies. “Passive” immunotherapy with monoclonal antibodies (mAbs) has outpaced “active” immunotherapy with anti-tumor vaccines, and mAbs that antagonize the off responses have been recently introduced in clinical practice. Despite these recent successes, many unresolved practical and theoretical questions remain. Notably unknown are the identity of the lymphocytes that eliminate tumor cells, which white cells enter into tumors, through which endothelium, in what order, and how they perform their task. The parameters of size and location that could be used to determine in which tumors the immune response may be sufficient to eradicate the tumor are yet unknown. Immunotherapy has been so far more efficient to treat solid and hematologic tumors located outside the central nervous system, than primary brain tumors and brain metastases. In contrast to recent advances with mAbs, anti-tumor vaccine development has been lagging behind. The multiplicity of antigens that must be targeted to achieve significant clinical response is partially responsible for this lag, especially in melanoma, one of the most mutated tumors. Further hampering vaccination results is the fact that tumor elimination by the immune system is the result of a race between tumors with different growth rates and the relatively slow development of the adaptive immune response. The enhancement of the native arm of the immune response or the administration of targeted chemotherapy to slow tumor development, are approaches that should be studied. Finally, criteria used to analyze patient response to immunotherapeutic treatments must be perfected, and the patient populations that could benefit the most from this approach must be better defined.
Collapse
Affiliation(s)
| | - Antonela Baron
- Laboratorio de Cancerología, Fundación Instituto Leloir - IIBBA-CONICET , Buenos Aires , Argentina
| | - Mariela Urrutia
- Laboratorio de Cancerología, Fundación Instituto Leloir - IIBBA-CONICET , Buenos Aires , Argentina
| | - José Mordoh
- Laboratorio de Cancerología, Fundación Instituto Leloir - IIBBA-CONICET , Buenos Aires , Argentina ; Centro de Investigaciones Oncológicas, Fundación Cáncer and Instituto Alexander Fleming , Buenos Aires , Argentina
| |
Collapse
|
694
|
Jarosz-Biej M, Smolarczyk R, Cichoń T, Kułach N, Czapla J, Matuszczak S, Szala S. Combined Tumor Cell-Based Vaccination and Interleukin-12 Gene Therapy Polarizes the Tumor Microenvironment in Mice. Arch Immunol Ther Exp (Warsz) 2015; 63:451-64. [PMID: 25801067 PMCID: PMC4633448 DOI: 10.1007/s00005-015-0337-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/09/2015] [Indexed: 12/22/2022]
Abstract
Tumor progression depends on tumor milieu, which influences neovasculature formation and immunosuppression. Combining immunotherapy with antiangiogenic/antivascular therapy might be an effective therapeutic approach. The aim of our study was to elaborate an anticancer therapeutic strategy based on the induction of immune response which leads to polarization of tumor milieu. To achieve this, we developed a tumor cell-based vaccine. CAMEL peptide was used as a B16-F10 cell death-inducing agent. The lysates were used as a vaccine to immunize mice bearing B16-F10 melanoma tumors. To further improve the therapeutic effect of the vaccine, we combined it with interleukin (IL)-12 gene therapy. IL-12, a cytokine with antiangiogenic properties, activates nonspecific and specific immune responses. We observed that combined therapy is significantly more effective (as compared with monotherapies) in inhibiting tumor growth. Furthermore, the tested combination polarizes the tumor microenvironment, which results in a switch from a proangiogenic/immunosuppressive to an antiangiogenic/immunostimulatory one. The switch manifests itself as a decreased number of tumor blood vessels, increased levels of tumor-infiltrating CD4+, CD8+ and NK cells, as well as lower level of suppressor lymphocytes (Treg). Our results suggest that polarizing tumor milieu by such combined therapy does inhibit tumor growth and seems to be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Magdalena Jarosz-Biej
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-101, Gliwice, Poland.
| | - Ryszard Smolarczyk
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-101, Gliwice, Poland
| | - Tomasz Cichoń
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-101, Gliwice, Poland
| | - Natalia Kułach
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-101, Gliwice, Poland
- Department of Animal Physiology and Ecotoxycology, University of Silesia, Katowice, Poland
| | - Justyna Czapla
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-101, Gliwice, Poland
| | - Sybilla Matuszczak
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-101, Gliwice, Poland
| | - Stanisław Szala
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15, 44-101, Gliwice, Poland
| |
Collapse
|
695
|
Kwilas AR, Donahue RN, Tsang KY, Hodge JW. Immune consequences of tyrosine kinase inhibitors that synergize with cancer immunotherapy. ACTA ACUST UNITED AC 2015; 2. [PMID: 26005708 DOI: 10.14800/ccm.677] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Combination therapy for the treatment of cancer is becoming increasingly essential as we gain improved understanding of the complexity of cancer progression and the mechanisms by which cancer cells become resistant to single-agent therapy. Recent studies, both clinical and preclinical, have suggested that immunotherapy is a promising approach to the treatment of cancer; however, strategies to improve its clinical efficacy are still needed. A number of recent studies have indicated that antiangiogenic tyrosine kinase inhibitors (TKIs) target multiple components of the tumor microenvironment and are an ideal class of agents for synergizing with cancer immunotherapy. TKIs are well known to modulate tumor endothelial cells, leading to vascular normalization; however, these agents have also been recently shown to decrease tumor compactness and tight junctions, thereby reducing solid tumor pressure and allowing for improved perfusion of collapsed vessels and increased tumor oxygenation. In addition, some TKIs are capable of inducing immunogenic modulation, whereby tumor cells are sensitized to killing by T lymphocytes. Moreover, a number of TKIs have been shown to be involved in immune subset conditioning, increasing the frequency and function of effector immune elements, while decreasing the number and function of immune suppressor cells. The alteration of the immune landscape, direct modification of tumor cells, and improved vascular perfusion leads to improved antitumor efficacy when antiangiogenic TKIs are combined with immunotherapy. Collectively, the data presented in this review support the clinical combination of multi-targeted antiangiogenic TKIs, including but not limited to cabozantinib, sunitinib, and sorafenib, as well as to other antiangiogenic therapies, such as the anti-VEGF antibody bevacizumab, with cancer vaccines for improved treatment of solid tumors.
Collapse
Affiliation(s)
- Anna R Kwilas
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Renee N Donahue
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kwong Y Tsang
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
696
|
Xu Y, Wang X. Fluid and cell behaviors along a 3D printed alginate/gelatin/fibrin channel. Biotechnol Bioeng 2015; 112:1683-95. [PMID: 25727058 DOI: 10.1002/bit.25579] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 02/17/2015] [Indexed: 02/06/2023]
Abstract
Three-dimensional (3D) cell manipulation is available with the integration of microfluidic technology and rapid prototyping techniques. High-Fidelity (Hi-Fi) constructs hold enormous therapeutic potential for organ manufacturing and regenerative medicine. In the present paper we introduced a quasi-three-dimensional (Q3D) model with parallel biocompatible alginate/gelatin/fibrin hurdles. The behaviors of fluids and cells along the microfluidic channels with various widths were studied. Cells inside the newly designed microfluidic channels attached and grew well. Morphological changes of adipose-derived stem cells (ADSCs) in both two-dimensional (2D) and 3D milieu were found on the printed constructs. Endothelialization occurred with the co-cultures of ADSCs and hepatocytes. This study provides insights into the interactions among fluids, cells and biomaterials, the behaviors of fluids and cells along the microfluidic channels, and the applications of Q3D techniques.
Collapse
Affiliation(s)
- Yufan Xu
- Department of Mechanical Engineering, Key Laboratory for Advanced Materials Processing Technology, Ministry of Education & Center of Organ Manufacturing, Tsinghua University, Beijing, 100084, P.R. China
| | - Xiaohong Wang
- Department of Mechanical Engineering, Key Laboratory for Advanced Materials Processing Technology, Ministry of Education & Center of Organ Manufacturing, Tsinghua University, Beijing, 100084, P.R. China. .,State Key Laboratory of Materials Processing and Die & Mould Technology, Huazhong University of Science and Technology, Wuhan, 430074, P.R. China.
| |
Collapse
|
697
|
Rivera LB, Bergers G. Intertwined regulation of angiogenesis and immunity by myeloid cells. Trends Immunol 2015; 36:240-9. [PMID: 25770923 DOI: 10.1016/j.it.2015.02.005] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 02/16/2015] [Accepted: 02/17/2015] [Indexed: 12/11/2022]
Abstract
Angiogenesis is a hallmark of cancer because its induction is indispensable to fuel an expanding tumor. The tumor microenvironment contributes to tumor vessel growth, and distinct myeloid cells recruited by the tumor have been shown not only to support angiogenesis but also to foster an immune suppressive environment that supports tumor expansion and progression. Recent findings suggest that the intertwined regulation of angiogenesis and immune modulation can offer therapeutic opportunities for the treatment of cancer. We review the mechanisms by which distinct myeloid cell populations contribute to tumor angiogenesis, discuss current approaches in the clinic that are targeting both angiogenic and immune suppressive pathways, and highlight important areas of future research.
Collapse
Affiliation(s)
- Lee B Rivera
- Department of Neurological Surgery, University of California, San Francisco, CA 94158, USA; University of California San Francisco (UCSF) Comprehensive Cancer Center, University of California, Helen Diller Family Cancer Research Center, San Francisco, CA 94158, USA.
| | - Gabriele Bergers
- Department of Neurological Surgery, University of California, San Francisco, CA 94158, USA; University of California San Francisco (UCSF) Comprehensive Cancer Center, University of California, Helen Diller Family Cancer Research Center, San Francisco, CA 94158, USA; Brain Tumor Center, University of California, San Francisco, CA 94158, USA.
| |
Collapse
|
698
|
Variations in genes involved in immune response checkpoints and association with outcomes in patients with resected colorectal liver metastases. THE PHARMACOGENOMICS JOURNAL 2015; 15:521-9. [PMID: 25752522 DOI: 10.1038/tpj.2015.14] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 12/05/2014] [Accepted: 01/28/2015] [Indexed: 01/10/2023]
Abstract
In patients with colorectal liver metastases (CLM), liver resection offers the possibility of cure and long-term survival. The liver is a highly immunogenic organ harboring ~80% of the body's tissue macrophages. Emerging data demonstrate a critical role of the immune response for cancer treatment. We investigated variations within genes involved in immune response checkpoints and their association with outcomes in patients with CLM who underwent neoadjuvant chemotherapy including bevacizumab and liver resection. Single-nucleotide polymorphisms (SNPs) in nine genes (CCL2, CCR2, LAG3, NT5E, PDCD1, CD274, IDO1, CTLA4 and CD24) were analyzed in genomic DNA from 149 patients with resected bevacizumab-pretreated CLM by direct Sanger DNA sequencing, and correlated with response, recurrence-free survival (RFS), overall survival (OS), probability of cure and recurrence patterns. IDO1 (indoleamine 2, 3-dioxygenase) rs3739319 G>A and CD24 rs8734 G>A showed a significant difference in 3-year OS rates. In addition, IDO1 rs3739319 G>A was significantly associated with extrahepatic recurrence. Recursive partitioning analyses revealed that IDO1 rs3739319 G>A was the dominant SNP predicting RFS and OS. Our data suggest that variants within genes involved in immune response checkpoints are associated with outcomes in patients with resected CLM and might lead to improved treatment strategies modulating anti-tumor immune response by targeting novel immune checkpoints.
Collapse
|
699
|
Zhang H, Ren Y, Tang X, Wang K, Liu Y, Zhang L, Li X, Liu P, Zhao C, He J. Vascular normalization induced by sinomenine hydrochloride results in suppressed mammary tumor growth and metastasis. Sci Rep 2015; 5:8888. [PMID: 25749075 PMCID: PMC4352869 DOI: 10.1038/srep08888] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 02/09/2015] [Indexed: 01/08/2023] Open
Abstract
Solid tumor vasculature is characterized by structural and functional abnormality and results in a hostile tumor microenvironment that mediates several deleterious aspects of tumor behavior. Sinomenine is an alkaloid extracted from the Chinese medicinal plant, Sinomenium acutum, which has been utilized to treat rheumatism in China for over 2000 years. Though sinomenine has been demonstrated to mediate a wide range of pharmacological actions, few studies have focused on its effect on tumor vasculature. We showed here that intraperitoneally administration of 100 mg/kg sinomenine hydrochloride (SH, the hydrochloride chemical form of sinomenine) in two orthotopic mouse breast cancer models for 14 days, delayed mammary tumor growth and decreased metastasis by inducing vascular maturity and enhancing tumor perfusion, while improving chemotherapy and tumor immunity. The effects of SH on tumor vessels were caused in part by its capability to restore the balance between pro-angiogenic factor (bFGF) and anti-angiogenic factor (PF4). However 200 mg/kg SH didn't exhibit the similar inhibitory effect on tumor progression due to the immunosuppressive microenvironment caused by excessive vessel pruning, G-CSF upregulation, and GM-CSF downregulation. Altogether, our findings suggest that SH induced vasculature normalization contributes to its anti-tumor and anti-metastasis effect on breast cancer at certain dosage.
Collapse
Affiliation(s)
- Huimin Zhang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, P.R. China
| | - Yu Ren
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, P.R. China
| | - Xiaojiang Tang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, P.R. China
| | - Ke Wang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, P.R. China
| | - Yang Liu
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, P.R. China
| | - Li Zhang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, P.R. China
| | - Xiao Li
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, P.R. China
| | - Peijun Liu
- Translational Medical center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, P.R. China
| | - Changqi Zhao
- Key Laboratory of Cell Proliferation and Regulation Biology, College of Life Science, Beijing Normal University, Beijing 100875, P.R. China
| | - Jianjun He
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an 710061, P.R. China
| |
Collapse
|
700
|
Chung TK, Warram J, Day KE, Hartman Y, Rosenthal EL. Time-dependent pretreatment with bevacuzimab increases tumor specific uptake of cetuximab in preclinical oral cavity cancer studies. Cancer Biol Ther 2015; 16:790-8. [PMID: 25719497 DOI: 10.1080/15384047.2015.1016664] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Inadequate delivery of therapeutics into tumors has been suggested as a reason for poor response. We hypothesize that bevacizumab, an antibody to vascular endothelial growth factor (VEGF), can improve cetuximab uptake in squamous cell carcinoma tumors. Athymic nude mice were implanted with OSC19 and SCC1 human cancer lines in a subcutaneous flank model. Mice were imaged daily for 14 days after intravenous tail vein injections of the following groups: IgG-IRDye800 (Control), cetuximab-IRDye800 (CTX800 Only), bevacizumab-IRDye800 (BVZ800 Only), cetuximab-IRDye800 + bevacuzimuab-IRDye800 (Simultaneous), and unlabeled bevacizumab followed by cetuximab-IRDye800 3 days later (Neoadjuvant). Within single-agent groups, the CTX800 Only tumor-specific uptake (TSU) was significantly higher than BVZ800 Only at Day 13 (TSU 8.6 vs 2.8, P < 0.001). The Simultaneous treatment with BVZ800 and CTX800 demonstrated no increase in antibody delivery. However, administration of unlabeled bevacizumab 3 days prior to CTX800 (Neoadjuvant group) resulted in significantly higher tumor specific delivery than administration of both antibodies at the same time (11.8 vs Simultaneous 5.0, P < 0.001). This difference can be attributed to a slower decline in tumor fluorescence intensity (-6.8% vs. Simultaneous -11.5% per day, respectively). Structural changes in pericyte coverage and functional vessel changes demonstrating decreased proliferation and tumor growth corroborate these fluorescence results. Although simultaneous administration of bevacizumab with cetuximab failed to increase antibody delivery to the tumor, pretreatment with bevacizumab improved TSU reflecting an increase in tumor-specific uptake of cetuximab as a result of vessel normalization.
Collapse
Affiliation(s)
- Thomas K Chung
- a Division of Otolaryngology; Department of Surgery; University of Alabama at Birmingham ; Birmingham , AL USA
| | | | | | | | | |
Collapse
|