51
|
Lu L, Saha D, Martuza RL, Rabkin SD, Wakimoto H. Single agent efficacy of the VEGFR kinase inhibitor axitinib in preclinical models of glioblastoma. J Neurooncol 2014; 121:91-100. [PMID: 25213669 DOI: 10.1007/s11060-014-1612-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 08/30/2014] [Indexed: 11/29/2022]
Abstract
Anti-angiogenic therapy is a promising therapeutic strategy for the highly vascular and malignant brain tumor, glioblastoma (GBM), although current clinical trials have failed to demonstrate an extension in overall survival. The small molecule tyrosine kinase inhibitor axitinib that targets vascular endothelial growth factor receptor, potently inhibits angiogenesis and has single-agent clinical activity in non-small cell lung, thyroid, and advanced renal cell cancer. Here we show that axitinib exerts direct cytotoxic activity against a number of patient-derived GBM stem cell (GSCs) and an endothelial cell line, and inhibits endothelial tube formation in vitro. Axitinib treatment of mice bearing hypervascular intracranial tumors generated from human U87 glioma cells, MGG4 GSCs and mouse 005 GSCs significantly extended survival that was associated with decreases in tumor-associated vascularity. We thus show for the first time the anti-angiogenic effect and survival prolongation provided by systemic single agent treatment with axitinib in preclinical orthotopic GBM models including clinically relevant GSC models. These results support further investigation of axitinib as an anti-angiogenic agent for GBM.
Collapse
Affiliation(s)
- Lei Lu
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | | | | | | | | |
Collapse
|
52
|
Intermittent metronomic drug schedule is essential for activating antitumor innate immunity and tumor xenograft regression. Neoplasia 2014; 16:84-96. [PMID: 24563621 DOI: 10.1593/neo.131910] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 12/24/2013] [Accepted: 01/02/2014] [Indexed: 12/24/2022] Open
Abstract
Metronomic chemotherapy using cyclophosphamide (CPA) is widely associated with antiangiogenesis; however, recent studies implicate other immune-based mechanisms, including antitumor innate immunity, which can induce major tumor regression in implanted brain tumor models. This study demonstrates the critical importance of drug schedule: CPA induced a potent antitumor innate immune response and tumor regression when administered intermittently on a 6-day repeating metronomic schedule but not with the same total exposure to activated CPA administered on an every 3-day schedule or using a daily oral regimen that serves as the basis for many clinical trials of metronomic chemotherapy. Notably, the more frequent metronomic CPA schedules abrogated the antitumor innate immune and therapeutic responses. Further, the innate immune response and antitumor activity both displayed an unusually steep dose-response curve and were not accompanied by antiangiogenesis. The strong recruitment of innate immune cells by the 6-day repeating CPA schedule was not sustained, and tumor regression was abolished, by a moderate (25%) reduction in CPA dose. Moreover, an ∼20% increase in CPA dose eliminated the partial tumor regression and weak innate immune cell recruitment seen in a subset of the every 6-day treated tumors. Thus, metronomic drug treatment must be at a sufficiently high dose but also sufficiently well spaced in time to induce strong sustained antitumor immune cell recruitment. Many current clinical metronomic chemotherapeutic protocols employ oral daily low-dose schedules that do not meet these requirements, suggesting that they may benefit from optimization designed to maximize antitumor immune responses.
Collapse
|
53
|
Kareva I. Immune evasion through competitive inhibition: the shielding effect of cancer non-stem cells. J Theor Biol 2014; 364:40-8. [PMID: 25195001 DOI: 10.1016/j.jtbi.2014.08.035] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 08/08/2014] [Accepted: 08/19/2014] [Indexed: 01/07/2023]
Abstract
It has been recently proposed that the two emerging hallmarks of cancer, namely altered glucose metabolism and immune evasion, may in fact be fundamentally linked. This connection comes from up-regulation of glycolysis by tumor cells, which can lead to active competition for resources in the tumor microenvironment between tumor and immune cells. Here it is further proposed that cancer stem cells (CSCs) can circumvent the anti-tumor immune response by creating a "protective shield" of non-stem cancer cells around them. This shield can protect the CSCs both by creating a physical barrier between them and cytotoxic lymphocytes (CTLs), and by promoting competition for the common resources, such as glucose, between non-stem cancer cells and CTLs. The implications of this hypothesis are investigated using an agent-based model, leading to a prediction that relative CSC to non-CSC ratio will vary depending on the strength of the host immune response. A discussion of possible therapeutic approaches concludes the paper, suggesting that a chemotherapeutic regimen consisting of regular pulsed doses, i.e., metronomic chemotherapy, would yield the best clinical outcome by removing the "protective shield" and thus allowing CTLs to most effectively reach and eliminate CSCs.
Collapse
Affiliation(s)
- Irina Kareva
- Newman Lakka Institute, Floating Hospital for Children at Tufts Medical Center, Boston, MA 02111, United States
| |
Collapse
|
54
|
Bruno A, Ferlazzo G, Albini A, Noonan DM. A think tank of TINK/TANKs: tumor-infiltrating/tumor-associated natural killer cells in tumor progression and angiogenesis. J Natl Cancer Inst 2014; 106:dju200. [PMID: 25178695 PMCID: PMC4344546 DOI: 10.1093/jnci/dju200] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Tumor-infiltrating leukocytes are often induced by the cancer microenvironment to display a protumor, proangiogenic phenotype. This “polarization” has been described for several myeloid cells, in particular macrophages. Natural killer (NK) cells represent another population of innate immune cells able to infiltrate tumors. The role of NK in tumor progression and angiogenesis has not yet been fully investigated. Several studies have shown that tumor-infiltrating NK (here referred to as “TINKs”) and tumor-associated NK (altered peripheral NK cells, which here we call “TANKs”) are compromised in their ability to lysew tumor cells. Recent data have suggested that they are potentially protumorigenic and can also acquire a proangiogenic phenotype. Here we review the properties of TINKs and TANKs and compare their activities to that of NK cells endowed with a physiological proangiogenic phenotype, in particular decidual NK cells. We speculate on the potential origins of TINKs and TANKs and on the immune signals involved in their differentiation and polarization. The TINK and TANK phenotype has broad implications in the immune response to tumors, ranging from a deficient control of cancer and cancer stem cells to an altered crosstalk with other relevant players of the immune response, such as dendritic cells, to induction of cancer angiogenesis. With this recently acquired knowledge that has not yet been put into perspective, we point out new potential avenues for therapeutic intervention involving NK cells as a target or an ally in oncology.
Collapse
Affiliation(s)
- Antonino Bruno
- Scientific and Technology Park, IRCCS MultiMedica, Milano, Italy (AB, DMN); Department of Human Pathology, University of Messina, Messina, Italy (GF); Department of Research and Statistics, IRCCS Arcispedale Santa Maria Nuova, Reggio Emilia, Italy (AA); Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy (DMN)
| | - Guido Ferlazzo
- Scientific and Technology Park, IRCCS MultiMedica, Milano, Italy (AB, DMN); Department of Human Pathology, University of Messina, Messina, Italy (GF); Department of Research and Statistics, IRCCS Arcispedale Santa Maria Nuova, Reggio Emilia, Italy (AA); Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy (DMN)
| | - Adriana Albini
- Scientific and Technology Park, IRCCS MultiMedica, Milano, Italy (AB, DMN); Department of Human Pathology, University of Messina, Messina, Italy (GF); Department of Research and Statistics, IRCCS Arcispedale Santa Maria Nuova, Reggio Emilia, Italy (AA); Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy (DMN)
| | - Douglas M Noonan
- Scientific and Technology Park, IRCCS MultiMedica, Milano, Italy (AB, DMN); Department of Human Pathology, University of Messina, Messina, Italy (GF); Department of Research and Statistics, IRCCS Arcispedale Santa Maria Nuova, Reggio Emilia, Italy (AA); Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy (DMN)
| |
Collapse
|
55
|
Wu J, Waxman DJ. Metronomic cyclophosphamide schedule-dependence of innate immune cell recruitment and tumor regression in an implanted glioma model. Cancer Lett 2014; 353:272-80. [PMID: 25069038 DOI: 10.1016/j.canlet.2014.07.033] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 07/17/2014] [Accepted: 07/19/2014] [Indexed: 01/13/2023]
Abstract
Metronomic cyclophosphamide (CPA) treatment activates robust innate anti-tumor immunity and induces major regression of large, implanted brain tumor xenografts when administered on an intermittent, every 6-day schedule, but not on a daily low-dose or a maximum-tolerated dose CPA schedule. Here, we used an implanted GL261 glioma model to compare five intermittent metronomic CPA schedules to elucidate the kinetics and schedule dependence of innate immune cell recruitment and tumor regression. Tumor-recruited natural killer cells induced by two every 6-day treatment cycles were significantly ablated 1 day after a third CPA treatment, but largely recovered several days later. Natural killer and other tumor-infiltrating innate immune cells peaked 12 days after the last CPA treatment on the every 6-day schedule, suggesting that drug-free intervals longer than 6 days may show increased efficacy. Metronomic CPA treatments spaced 9 or 12 days apart, or on an alternating 6 and 9 day schedule, induced extensive tumor regression, similar to the 6-day schedule; however, the tumor-infiltrating natural killer cell responses were not sustained, leading to rapid resumption of tumor growth after day 24, despite ongoing metronomic CPA treatment. Increasing the CPA dose prolonged the period of tumor regression on the every 9-day schedule, but natural killer cell activation was markedly decreased. Thus, while several intermittent metronomic CPA treatment schedules can activate innate immune cell recruitment leading to major tumor regression, sustained immune and anti-tumor responses are only achieved on the 6-day schedule. However, even with this schedule, some tumors eventually relapse, indicating a need for further improvements in this immunogenic metronomic therapy.
Collapse
Affiliation(s)
- Junjie Wu
- Division of Cell and Molecular Biology, Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - David J Waxman
- Division of Cell and Molecular Biology, Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA.
| |
Collapse
|
56
|
Li M, Bolduc AR, Hoda MN, Gamble DN, Dolisca SB, Bolduc AK, Hoang K, Ashley C, McCall D, Rojiani AM, Maria BL, Rixe O, MacDonald TJ, Heeger PS, Mellor AL, Munn DH, Johnson TS. The indoleamine 2,3-dioxygenase pathway controls complement-dependent enhancement of chemo-radiation therapy against murine glioblastoma. J Immunother Cancer 2014; 2:21. [PMID: 25054064 PMCID: PMC4105871 DOI: 10.1186/2051-1426-2-21] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 07/02/2014] [Indexed: 12/31/2022] Open
Abstract
Background Indoleamine 2,3-dioxygenase (IDO) is an enzyme with immune-suppressive properties that is commonly exploited by tumors to evade immune destruction. Anti-tumor T cell responses can be initiated in solid tumors, but are immediately suppressed by compensatory upregulation of immunological checkpoints, including IDO. In addition to these known effects on the adaptive immune system, we previously showed widespread, T cell-dependent complement deposition during allogeneic fetal rejection upon maternal treatment with IDO-blockade. We hypothesized that IDO protects glioblastoma from the full effects of chemo-radiation therapy by preventing vascular activation and complement-dependent tumor destruction. Methods To test this hypothesis, we utilized a syngeneic orthotopic glioblastoma model in which GL261 glioblastoma tumor cells were stereotactically implanted into the right frontal lobes of syngeneic mice. These mice were treated with IDO-blocking drugs in combination with chemotherapy and radiation therapy. Results Pharmacologic inhibition of IDO synergized with chemo-radiation therapy to prolong survival in mice bearing intracranial glioblastoma tumors. We now show that pharmacologic or genetic inhibition of IDO allowed chemo-radiation to trigger widespread complement deposition at sites of tumor growth. Chemotherapy treatment alone resulted in collections of perivascular leukocytes within tumors, but no complement deposition. Adding IDO-blockade led to upregulation of VCAM-1 on vascular endothelium within the tumor microenvironment, and further adding radiation in the presence of IDO-blockade led to widespread deposition of complement. Mice genetically deficient in complement component C3 lost all of the synergistic effects of IDO-blockade on chemo-radiation-induced survival. Conclusions Together these findings identify a novel mechanistic link between IDO and complement, and implicate complement as a major downstream effector mechanism for the beneficial effect of IDO-blockade after chemo-radiation therapy. We speculate that this represents a fundamental pathway by which the tumor regulates intratumoral vascular activation and protects itself from immune-mediated tumor destruction.
Collapse
Affiliation(s)
- Minghui Li
- GRU Cancer Center, Georgia Regents University, Augusta, Georgia, 30912, USA ; Program in Cancer immunology, Inflammation and Tolerance (CIT), Georgia Regents University, Augusta, GA, USA ; Medical College of Georgia Department of Pediatrics, Georgia Regents University, 1120 Fifteenth Street, Augusta, GA CN-4141A, USA
| | - Aaron R Bolduc
- GRU Cancer Center, Georgia Regents University, Augusta, Georgia, 30912, USA ; Program in Cancer immunology, Inflammation and Tolerance (CIT), Georgia Regents University, Augusta, GA, USA ; Department of Surgery, Georgia Regents University, Augusta, GA, USA
| | - Md Nasrul Hoda
- Department of Neurology, Georgia Regents University, Augusta, GA, USA ; College of Allied Health Sciences Department of Medical Laboratory, Imaging & Radiologic Sciences, Georgia Regents University, Augusta, GA 30912, USA
| | - Denise N Gamble
- GRU Cancer Center, Georgia Regents University, Augusta, Georgia, 30912, USA ; Program in Cancer immunology, Inflammation and Tolerance (CIT), Georgia Regents University, Augusta, GA, USA
| | - Sarah-Bianca Dolisca
- Medical College of Georgia Department of Pediatrics, Georgia Regents University, 1120 Fifteenth Street, Augusta, GA CN-4141A, USA
| | - Anna K Bolduc
- GRU Cancer Center, Georgia Regents University, Augusta, Georgia, 30912, USA ; Program in Cancer immunology, Inflammation and Tolerance (CIT), Georgia Regents University, Augusta, GA, USA
| | - Kelly Hoang
- Medical College of Georgia Department of Pediatrics, Georgia Regents University, 1120 Fifteenth Street, Augusta, GA CN-4141A, USA
| | - Claire Ashley
- Medical College of Georgia Department of Pediatrics, Georgia Regents University, 1120 Fifteenth Street, Augusta, GA CN-4141A, USA
| | - David McCall
- GRU Cancer Center, Georgia Regents University, Augusta, Georgia, 30912, USA ; Program in Cancer immunology, Inflammation and Tolerance (CIT), Georgia Regents University, Augusta, GA, USA
| | - Amyn M Rojiani
- GRU Cancer Center, Georgia Regents University, Augusta, Georgia, 30912, USA ; Department of Pathology, Georgia Regents University, Augusta, GA, USA
| | - Bernard L Maria
- GRU Cancer Center, Georgia Regents University, Augusta, Georgia, 30912, USA ; Medical College of Georgia Department of Pediatrics, Georgia Regents University, 1120 Fifteenth Street, Augusta, GA CN-4141A, USA ; Department of Neurology, Georgia Regents University, Augusta, GA, USA ; Department of Neurosurgery, Georgia Regents University, Augusta, GA, USA
| | - Olivier Rixe
- GRU Cancer Center, Georgia Regents University, Augusta, Georgia, 30912, USA ; Department of Medicine, Georgia Regents University, Augusta, GA, USA
| | - Tobey J MacDonald
- Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Peter S Heeger
- Department of Medicine, Division of Nephrology, The Immunology Institute, New York, NY 10025, USA ; Recanati-Miller Transplant Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10025, USA
| | - Andrew L Mellor
- GRU Cancer Center, Georgia Regents University, Augusta, Georgia, 30912, USA ; Program in Cancer immunology, Inflammation and Tolerance (CIT), Georgia Regents University, Augusta, GA, USA ; Department of Medicine, Georgia Regents University, Augusta, GA, USA
| | - David H Munn
- GRU Cancer Center, Georgia Regents University, Augusta, Georgia, 30912, USA ; Program in Cancer immunology, Inflammation and Tolerance (CIT), Georgia Regents University, Augusta, GA, USA ; Medical College of Georgia Department of Pediatrics, Georgia Regents University, 1120 Fifteenth Street, Augusta, GA CN-4141A, USA
| | - Theodore S Johnson
- GRU Cancer Center, Georgia Regents University, Augusta, Georgia, 30912, USA ; Program in Cancer immunology, Inflammation and Tolerance (CIT), Georgia Regents University, Augusta, GA, USA ; Medical College of Georgia Department of Pediatrics, Georgia Regents University, 1120 Fifteenth Street, Augusta, GA CN-4141A, USA
| |
Collapse
|
57
|
Ajaz M, Jefferies S, Brazil L, Watts C, Chalmers A. Current and investigational drug strategies for glioblastoma. Clin Oncol (R Coll Radiol) 2014; 26:419-30. [PMID: 24768122 DOI: 10.1016/j.clon.2014.03.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 03/27/2014] [Indexed: 11/21/2022]
Abstract
Medical treatments for glioblastoma face several challenges. Lipophilic alkylators remain the mainstay of treatment, emphasising the primacy of good blood-brain barrier penetration. Temozolomide has emerged as a major contributor to improved patient survival. The roles of procarbazine and vincristine in the procarbazine, lomustine and vincristine (PCV) schedule have attracted scrutiny and several lines of evidence now support the use of lomustine as effective single-agent therapy. Bevacizumab has had a convoluted development history, but clearly now has no major role in first-line treatment, and may even be detrimental to quality of life in this setting. In later disease, clinically meaningful benefits are achievable in some patients, but more impressively the combination of bevacizumab and lomustine shows early promise. Over the last decade, investigational strategies in glioblastoma have largely subscribed to the targeted kinase inhibitor paradigm and have mostly failed. Low prevalence dominant driver lesions such as the FGFR-TACC fusion may represent a niche role for this agent class. Immunological, metabolic and radiosensitising approaches are being pursued and offer more generalised efficacy. Finally, trial design is a crucial consideration. Progress in clinical glioblastoma research would be greatly facilitated by improved methodologies incorporating: (i) routine pharmacokinetic and pharmacodynamic assessments by preoperative dosing; and (ii) multi-stage, multi-arm protocols incorporating new therapy approaches and high-resolution biology in order to guide necessary improvements in science.
Collapse
Affiliation(s)
- M Ajaz
- Surrey Cancer Research Institute, University of Surrey, Guildford, UK.
| | - S Jefferies
- Oncology Centre, Addenbrooke's Hospital, Cambridge, UK
| | - L Brazil
- Guy's, St Thomas' and King's College Hospitals, London, UK
| | - C Watts
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - A Chalmers
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
58
|
Doloff JC, Chen CS, Waxman DJ. Anti-tumor innate immunity activated by intermittent metronomic cyclophosphamide treatment of 9L brain tumor xenografts is preserved by anti-angiogenic drugs that spare VEGF receptor 2. Mol Cancer 2014; 13:158. [PMID: 24965046 PMCID: PMC4083145 DOI: 10.1186/1476-4598-13-158] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 06/16/2014] [Indexed: 01/04/2023] Open
Abstract
Background Metronomic cyclophosphamide given on an intermittent, 6-day repeating schedule, but not on an exposure dose-equivalent daily schedule, activates an anti-tumor innate immune response that leads to major regression of large implanted gliomas, without anti-angiogenesis. Methods and approach Mice bearing implanted 9L gliomas were used to investigate the effects of this 6-day repeating, immunogenic cyclophosphamide schedule on myeloid-derived suppressor cells, which are pro-angiogenic and can inhibit anti-tumor immunity, and to elucidate the mechanism whereby the innate immune cell-dependent tumor regression response to metronomic cyclophosphamide treatment is blocked by several anti-angiogenic receptor tyrosine kinase inhibitors. Results Intermittent metronomic cyclophosphamide scheduling strongly increased glioma-associated CD11b+ immune cells but not CD11b+Gr1+ myeloid-derived suppressor cells, while bone marrow and spleen reservoirs of the suppressor cells were decreased. The inhibition of immune cell recruitment and tumor regression by anti-angiogenic receptor tyrosine kinase inhibitors, previously observed in several brain tumor models, was recapitulated in the 9L tumor model with the VEGFR2-specific inhibitory monoclonal antibody DC101 (p < 0.01), implicating VEGFR2 signaling as an essential step in metronomic cyclophosphamide-stimulated immune cell recruitment. In contrast, sorafenib, a multi-receptor tyrosine kinase inhibitor with comparatively weak VEGF receptor phosphorylation inhibitory activity, was strongly anti-angiogenic but did not block metronomic cyclophosphamide-induced innate immunity or tumor regression (p > 0.05). Conclusions The interference by receptor tyrosine kinase inhibitors in the immunogenic actions of intermittent metronomic chemotherapy is not a consequence of anti-angiogenesis per se, as demonstrated in an implanted 9L tumor model. Furthermore, this undesirable interaction with tyrosine kinase inhibitors can be avoided by using anti-angiogenic drugs that spare the VEGFR2 pathway.
Collapse
Affiliation(s)
| | | | - David J Waxman
- Division of Cell and Molecular Biology, Department of Biology, Boston University, 5 Cummington Street, Boston, MA 02215, USA.
| |
Collapse
|
59
|
Hasnis E, Alishekevitz D, Gingis-Veltski S, Bril R, Fremder E, Voloshin T, Raviv Z, Karban A, Shaked Y. Anti-Bv8 antibody and metronomic gemcitabine improve pancreatic adenocarcinoma treatment outcome following weekly gemcitabine therapy. Neoplasia 2014; 16:501-10. [PMID: 24957319 PMCID: PMC4198746 DOI: 10.1016/j.neo.2014.05.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 05/27/2014] [Accepted: 05/30/2014] [Indexed: 12/13/2022] Open
Abstract
Weekly gemcitabine therapy is the major treatment offered for patients with pancreatic adenocarcinoma cancer; however, relative resistance of tumor cells to chemotherapy, rapid regrowth, and metastasis are the main causes of death within a year. Recently, the daily continuous administration of chemotherapy in low doses--called metronomic chemotherapy (MC)--has been shown to inhibit primary tumor growth and delay metastases in several tumor types; however, its use as a single therapy is still in question due to its moderate therapeutic benefit. Here, we show that the combination of weekly gemcitabine with MC of the same drug delays tumor regrowth and inhibits metastasis in mice implanted orthotopically with pancreatic tumors. We further demonstrate that weekly gemcitabine, but not continuous MC gemcitabine or the combination of the two drug regimens, promotes rebound myeloid-derived suppressor cell (MDSC) mobilization and increases angiogenesis in this tumor model. Furthermore, Bv8 is highly expressed in MDSCs colonizing pancreatic tumors in mice treated with weekly gemcitabine compared to MC gemcitabine or the combination of the two regimens. Blocking Bv8 with antibodies in weekly gemcitabine-treated mice results in a significant reduction in tumor regrowth, angiogenesis, and metastasis. Overall, our results suggest that pro-tumorigenic effects induced by weekly gemcitabine are mediated in part by MDSCs expressing Bv8. Therefore, both Bv8 inhibition and MC can be used as legitimate 'add-on' treatments for preventing post-chemotherapy pancreatic cancer recurrence, progression, and metastasis following weekly gemcitabine therapy.
Collapse
Affiliation(s)
- Erez Hasnis
- Department of Molecular Pharmacology, Rappaport Faculty of Medicine, Technion, Haifa, Israel; Department of Internal Medicine C, Rambam Health Care Campus, Haifa, Israel
| | - Dror Alishekevitz
- Department of Molecular Pharmacology, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Svetlana Gingis-Veltski
- Department of Molecular Pharmacology, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Rotem Bril
- Department of Molecular Pharmacology, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Ella Fremder
- Department of Molecular Pharmacology, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Tali Voloshin
- Department of Molecular Pharmacology, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Ziv Raviv
- Department of Molecular Pharmacology, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Amir Karban
- Department of Internal Medicine C, Rambam Health Care Campus, Haifa, Israel
| | - Yuval Shaked
- Department of Molecular Pharmacology, Rappaport Faculty of Medicine, Technion, Haifa, Israel.
| |
Collapse
|
60
|
Chretien AS, Le Roy A, Vey N, Prebet T, Blaise D, Fauriat C, Olive D. Cancer-Induced Alterations of NK-Mediated Target Recognition: Current and Investigational Pharmacological Strategies Aiming at Restoring NK-Mediated Anti-Tumor Activity. Front Immunol 2014; 5:122. [PMID: 24715892 PMCID: PMC3970020 DOI: 10.3389/fimmu.2014.00122] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 03/10/2014] [Indexed: 01/14/2023] Open
Abstract
Despite evidence of cancer immune-surveillance, which plays a key role in tumor rejection, cancer cells can escape immune recognition through different mechanisms. Thus, evasion to Natural killer (NK) cell-mediated anti-tumor activity is commonly described and is mediated by various mechanisms, mainly cancer cell-induced down-regulation of NK-activating receptors (NCRs, NKG2D, DNAM-1, and CD16) as well as up-regulation of inhibitory receptors (killer-cell immunoglobulin-like receptors, KIRs, NKG2A). Alterations of NK cells lead to an impaired recognition of tumor cells as well as a decreased ability to interact with immune cells. Alternatively, cancer cells downregulate expression of ligands for NK cell-activating receptors and up-regulate expression of the ligands for inhibitory receptors. A better knowledge of the extent and the mechanisms of these defects will allow developing pharmacological strategies to restore NK cell ability to recognize and lyse tumor cells. Combining conventional chemotherapy and immune modulation is a promising approach likely to improve clinical outcome in diverse neoplastic malignancies. Here, we overview experimental approaches as well as strategies already available in the clinics that restore NK cell functionality. Yet successful cancer therapies based on the manipulation of NK cell already have shown efficacy in the context of hematologic malignancies. Additionally, the ability of cytotoxic agents to increase susceptibility of tumors to NK cell lysis has been studied and may require improvement to maximize this effect. More recently, new strategies were developed to specifically restore NK cell phenotype or to stimulate NK cell functions. Overall, pharmacological immune modulation trends to be integrated in therapeutic strategies and should improve anti-tumor effects of conventional cancer therapy.
Collapse
Affiliation(s)
- Anne-Sophie Chretien
- Centre de Cancérologie de Marseille, INSERM, U1068, Institut Paoli-Calmettes, Aix-Marseille Université, UM 105, CNRS, UMR7258, Marseille, France
| | - Aude Le Roy
- Centre de Cancérologie de Marseille, Plateforme d’Immunomonitoring en Cancérologie, INSERM, U1068, Institut Paoli-Calmettes, Aix-Marseille Université, UM 105, CNRS, UMR7258, Marseille, France
| | - Norbert Vey
- Centre de Cancérologie de Marseille, INSERM, U1068, Institut Paoli-Calmettes, Aix-Marseille Université, UM 105, CNRS, UMR7258, Marseille, France
- Département d’Hématologie, Institut Paoli-Calmettes, Marseille, France
| | - Thomas Prebet
- Département d’Hématologie, Institut Paoli-Calmettes, Marseille, France
| | - Didier Blaise
- Centre de Cancérologie de Marseille, INSERM, U1068, Institut Paoli-Calmettes, Aix-Marseille Université, UM 105, CNRS, UMR7258, Marseille, France
- Unité de Transplantation et de Thérapie Cellulaire, Institut Paoli-Calmettes, Marseille, France
| | - Cyril Fauriat
- Centre de Cancérologie de Marseille, INSERM, U1068, Institut Paoli-Calmettes, Aix-Marseille Université, UM 105, CNRS, UMR7258, Marseille, France
| | - Daniel Olive
- Centre de Cancérologie de Marseille, INSERM, U1068, Institut Paoli-Calmettes, Aix-Marseille Université, UM 105, CNRS, UMR7258, Marseille, France
- Centre de Cancérologie de Marseille, Plateforme d’Immunomonitoring en Cancérologie, INSERM, U1068, Institut Paoli-Calmettes, Aix-Marseille Université, UM 105, CNRS, UMR7258, Marseille, France
| |
Collapse
|
61
|
Koumarianou A, Christodoulou MI, Patapis P, Papadopoulos I, Liakata E, Giagini A, Stavropoulou A, Poulakaki N, Tountas N, Xiros N, Economopoulos T, Pectasides D, Tsitsilonis OE, Pappa V. The effect of metronomic versus standard chemotherapy on the regulatory to effector T-cell equilibrium in cancer patients. Exp Hematol Oncol 2014; 3:3. [PMID: 24456704 PMCID: PMC3906764 DOI: 10.1186/2162-3619-3-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 01/11/2014] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The host's immune system is crucially involved in cancer development and progression. The ratio of regulatory to effector T-cells, as well as the interplay of T-cells with therapeutic agents, impact on cancer prognosis. The current study aimed to comparatively investigate the effect of metronomic and standard chemotherapy on the number and functionality of peripheral regulatory and effector T-cells in cancer patients. METHODS CD4+CD25+ regulatory and CD4+CD25- effector T-cells were purified from the peripheral blood of 36 cancer patients and co-cultured in the presence of a polyclonal stimulus. The proliferative capacity and frequency of CD4+CD25+/CD4+CD25- T-cells were analysed before and during various chemotherapeutic regimes, by ELISA and flow cytometry, respectively. RESULTS Chemotherapy shifted immune responses in favour of regulatory T-cells. The relative ratio of regulatory to effector T-cells increased, and the T-cell-mediated suppressive activity of regulatory on effector T-cells was augmented. This effect was more profound in metronomic than in standard chemotherapeutic approaches. Moreover, an association between the chemotherapy strategy followed and the mode of action of specific drugs (anti-mitotic, anti-DNA) was revealed. CONCLUSIONS In comparison to standard chemotherapeutic strategies, metronomic approaches, though more patient-friendly, result in a significantly more prominent expansion of regulatory T-cells that aggravate the regulatory to effector T-cell imbalance. Our findings impact on the modulation of chemotherapy-treated patients' anti-tumor immunity and, thus, may be proven useful for selecting the most advantageous drug-delivery strategy, particularly when immunotherapeutics are eventually to be applied.
Collapse
Affiliation(s)
- Anna Koumarianou
- Fourth Department of Internal Medicine, Attikon University Hospital, Rimini 1 Street, 12462 Athens, Greece.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
|
63
|
Bracci L, Schiavoni G, Sistigu A, Belardelli F. Immune-based mechanisms of cytotoxic chemotherapy: implications for the design of novel and rationale-based combined treatments against cancer. Cell Death Differ 2014; 21:15-25. [PMID: 23787994 PMCID: PMC3857622 DOI: 10.1038/cdd.2013.67] [Citation(s) in RCA: 689] [Impact Index Per Article: 62.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 05/07/2013] [Accepted: 05/14/2013] [Indexed: 02/06/2023] Open
Abstract
Conventional anticancer chemotherapy has been historically thought to act through direct killing of tumor cells. This concept stems from the fact that cytotoxic drugs interfere with DNA synthesis and replication. Accumulating evidence, however, indicates that the antitumor activities of chemotherapy also rely on several off-target effects, especially directed to the host immune system, that cooperate for successful tumor eradication. Chemotherapeutic agents stimulate both the innate and adaptive arms of the immune system through several modalities: (i) by promoting specific rearrangements on dying tumor cells, which render them visible to the immune system; (ii) by influencing the homeostasis of the hematopoietic compartment through transient lymphodepletion followed by rebound replenishment of immune cell pools; (iii) by subverting tumor-induced immunosuppressive mechanisms and (iv) by exerting direct or indirect stimulatory effects on immune effectors. Among the indirect ways of immune cell stimulation, some cytotoxic drugs have been shown to induce an immunogenic type of cell death in tumor cells, resulting in the emission of specific signals that trigger phagocytosis of cell debris and promote the maturation of dendritic cells, ultimately resulting in the induction of potent antitumor responses. Here, we provide an extensive overview of the multiple immune-based mechanisms exploited by the most commonly employed cytotoxic drugs, with the final aim of identifying prerequisites for optimal combination with immunotherapy strategies for the development of more effective treatments against cancer.
Collapse
Affiliation(s)
- L Bracci
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - G Schiavoni
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - A Sistigu
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - F Belardelli
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
64
|
Kelley RK, Hwang J, Magbanua MJM, Watt L, Beumer JH, Christner SM, Baruchel S, Wu B, Fong L, Yeh BM, Moore AP, Ko AH, Korn WM, Rajpal S, Park JW, Tempero MA, Venook AP, Bergsland EK. A phase 1 trial of imatinib, bevacizumab, and metronomic cyclophosphamide in advanced colorectal cancer. Br J Cancer 2013; 109:1725-34. [PMID: 24022191 PMCID: PMC3790192 DOI: 10.1038/bjc.2013.553] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 07/14/2013] [Accepted: 08/19/2013] [Indexed: 02/08/2023] Open
Abstract
Background: This phase 1 clinical trial was conducted to determine the safety, maximum-tolerated dose (MTD), and pharmacokinetics of imatinib, bevacizumab, and metronomic cyclophosphamide in patients with advanced colorectal cancer (CRC). Methods: Patients with refractory stage IV CRC were treated with bevacizumab 5 mg kg−1 i.v. every 2 weeks (fixed dose) plus oral cyclophosphamide q.d. and imatinib q.d. or b.i.d. in 28-day cycles with 3+3 dose escalation. Response was assessed every two cycles. Pharmacokinetics of imatinib and cyclophosphamide and circulating tumour, endothelial, and immune cell subsets were measured. Results: Thirty-five patients were enrolled. Maximum-tolerated doses were cyclophosphamide 50 mg q.d., imatinib 400 mg q.d., and bevacizumab 5 mg kg−1 i.v. every 2 weeks. Dose-limiting toxicities (DLTs) included nausea/vomiting, neutropaenia, hyponatraemia, fistula, and haematuria. The DLT window required expansion to 42 days (1.5 cycles) to capture delayed toxicities. Imatinib exposure increased insignificantly after adding cyclophosphamide. Seven patients (20%) experienced stable disease for >6 months. Circulating tumour, endothelial, or immune cells were not associated with progression-free survival. Conclusion: The combination of metronomic cyclophosphamide, imatinib, and bevacizumab is safe and tolerable without significant drug interactions. A subset of patients experienced prolonged stable disease independent of dose level.
Collapse
Affiliation(s)
- R K Kelley
- Department of Medicine, University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, 1600 Divisadero Street, Box 1700, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Hahnfeldt P, Hlatky L, Klement GL. Center of cancer systems biology second annual workshop--tumor metronomics: timing and dose level dynamics. Cancer Res 2013; 73:2949-54. [PMID: 23492368 DOI: 10.1158/0008-5472.can-12-3807] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Metronomic chemotherapy, the delivery of doses in a low, regular manner so as to avoid toxic side effects, was introduced over 12 years ago in the face of substantial clinical and preclinical evidence supporting its tumor-suppressive capability. It constituted a marked departure from the classic maximum-tolerated dose (MTD) strategy, which, given its goal of rapid eradication, uses dosing sufficiently intense to require rest periods between cycles to limit toxicity. Even so, upfront tumor eradication is frequently not achieved with MTD, whereupon a de facto goal of longer-term tumor control is often pursued. As metronomic dosing has shown tumor control capability, even for cancers that have become resistant to the same drug delivered under MTD, the question arises whether it may be a preferable alternative dosing approach from the outset. To date, however, our knowledge of the coupled dynamics underlying metronomic dosing is neither sufficiently well developed nor widely enough disseminated to establish its actual potential. Meeting organizers thus felt the time was right, armed with new quantitative approaches, to call a workshop on "Tumor Metronomics: Timing and Dose Level Dynamics" to explore prospects for gaining a deeper, systems-level appreciation of the metronomics concept. The workshop proved to be a forum in which experts from the clinical, biologic, mathematical, and computational realms could work together to clarify the principles and underpinnings of metronomics. Among other things, the need for significant shifts in thinking regarding endpoints to be used as clinical standards of therapeutic progress was recognized.
Collapse
Affiliation(s)
- Philip Hahnfeldt
- Center of Cancer Systems Biology, Steward St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA, USA.
| | | | | |
Collapse
|
66
|
Manley E, Waxman DJ. Impact of tumor blood flow modulation on tumor sensitivity to the bioreductive drug banoxantrone. J Pharmacol Exp Ther 2013; 344:368-77. [PMID: 23192656 PMCID: PMC3558827 DOI: 10.1124/jpet.112.200089] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 11/27/2012] [Indexed: 12/17/2022] Open
Abstract
We investigated the hypoxia-dependent cytotoxicity of AQ4N (banoxantrone) using a panel of 13 cancer cell lines and studied its relationship to the expression of the quinone reductase DT-diaphorase (NQO1), which is widely found in cancer cells. We also investigated pharmacologic treatments that increase tumor hypoxia in vivo and their impact on AQ4N chemosensitivity in a solid tumor xenograft model. AQ4N showed ≥ 8-fold higher cytotoxicity under hypoxia than normoxia in cultures of 9L rat gliosarcoma and H460 human non-small-cell lung carcinoma cells but not for 11 other human cancer cell lines. DT-diaphorase protein levels and AQ4N chemosensitivity were poorly correlated across the cancer cell line panel, and AQ4N chemosensitivity was not affected by DT-diaphorase inhibitors. The vasodilator hydralazine decreased tumor perfusion and increased tumor hypoxia in 9L tumor xenografts, and to a lesser extent in H460 tumor xenografts. However, hydralazine did not increase AQ4N-dependent antitumor activity. Combination of AQ4N with the angiogenesis inhibitor axitinib, which increases 9L tumor hypoxia, transiently increased antitumor activity but with an increase in host toxicity. These findings indicate that the capacity to bioactivate AQ4N is not dependent on DT-diaphorase and is not widespread in cultured cancer cell lines. Moreover, the activation of AQ4N cytotoxicity in vivo requires tumor hypoxia that is more extensive or prolonged than can readily be achieved by vasodilation or by antiangiogenic drug treatment.
Collapse
Affiliation(s)
- Eugene Manley
- Department of Biology, Boston University, 5 Cummington Street, Boston, MA 02215, USA
| | | |
Collapse
|
67
|
Low-dose metronomic chemotherapy: from past experience to new paradigms in the treatment of cancer. Drug Discov Today 2013; 18:193-201. [PMID: 22868084 DOI: 10.1016/j.drudis.2012.07.015] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 06/28/2012] [Accepted: 07/26/2012] [Indexed: 01/22/2023]
|
68
|
Jia L, Waxman DJ. Thrombospondin-1 and pigment epithelium-derived factor enhance responsiveness of KM12 colon tumor to metronomic cyclophosphamide but have disparate effects on tumor metastasis. Cancer Lett 2012; 330:241-9. [PMID: 23228633 DOI: 10.1016/j.canlet.2012.11.055] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 11/30/2012] [Accepted: 11/30/2012] [Indexed: 12/18/2022]
Abstract
The anti-tumor activity, metronomic chemotherapy sensitization potential and metastatic effects of the endogenous angiogenesis inhibitors thrombospondin-1 and PEDF were investigated in KM12 colon adenocarcinoma xenografts. Thrombospondin-1 and PEDF decreased KM12 tumor microvessel density, increased macrophage infiltration, and improved responsiveness to metronomic cyclophosphamide (CPA) treatment, but did not activate the anti-tumor innate immunity that metronomic CPA induces in other tumor models. Moreover, thrombospondin-1, but not PEDF, significantly increased KM12 metastasis to the lung, while PEDF augmented the anti-metastatic activity of metronomic CPA. Thus, while thrombospondin-1 and PEDF both increase the KM12 tumor responsiveness to metronomic CPA, they have disparate effects on tumor metastasis.
Collapse
Affiliation(s)
- Li Jia
- Division of Cell and Molecular Biology, Department of Biology, Boston University, Boston, MA 02215, United States
| | | |
Collapse
|
69
|
Mross K, Steinbild S. Metronomic anti-cancer therapy – an ongoing treatment option for advanced cancer patients. ACTA ACUST UNITED AC 2012. [DOI: 10.7243/2049-7962-1-32] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|